Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Journal of

Personalized
Medicine

Review
Cerebral Oxygen Delivery and Consumption in Brain-Injured
Patients
Dorota Siwicka-Gieroba 1, * , Chiara Robba 2,3 , Jakub Gołacki 1 , Rafael Badenes 4 and Wojciech Dabrowski 1

1 Department of Anaesthesiology and Intensive Care, Medical University in Lublin, 20-954 Lublin, Poland
2 Department of Anesthesiology and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology
and Neurosciences, 16132 Genoa, Italy
3 Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
4 Department of Anesthesiology and Surgical-Trauma Intensive Care, Hospital Clinic Universitari,
University of Valencia, 46010 Valencia, Spain
* Correspondence: dsiw@wp.pl

Abstract: Organism survival depends on oxygen delivery and utilization to maintain the balance of
energy and toxic oxidants production. This regulation is crucial to the brain, especially after acute
injuries. Secondary insults after brain damage may include impaired cerebral metabolism, ischemia,
intracranial hypertension and oxygen concentration disturbances such as hypoxia or hyperoxia.
Recent data highlight the important role of clinical protocols in improving oxygen delivery and
resulting in lower mortality in brain-injured patients. Clinical protocols guide the rules for oxygen
supplementation based on physiological processes such as elevation of oxygen supply (by mean
arterial pressure (MAP) and intracranial pressure (ICP) modulation, cerebral vasoreactivity, oxygen
capacity) and reduction of oxygen demand (by pharmacological sedation and coma or hypothermia).
The aim of this review is to discuss oxygen metabolism in the brain under different conditions.
Citation: Siwicka-Gieroba, D.; Robba,
C.; Gołacki, J.; Badenes, R.;
Keywords: brain; oxygen; delivery; consumption; TBI
Dabrowski, W. Cerebral Oxygen
Delivery and Consumption in
Brain-Injured Patients. J. Pers. Med.
2022, 12, 1763. https://doi.org/ 1. Introduction
10.3390/jpm12111763 Organism survival depends on oxygen delivery and utilization to maintain the balance
Academic Editors: Manuel of energy and toxic oxidants production [1]. This regulation is crucial to the central
V. Granell-Gil, Juan P. Cata and nervous system (CNS). Brain tissue presents a peculiarly dynamic consumption of energy.
Eduardo Tamayo The most productive metabolic process of energy analogs is oxidative phosphorylation,
relating to oxygen consumption [2]. Already in 1890, Roy and Sherrington observed that
Received: 27 August 2022
increased neuronal activity elevates energy consumption and compensatory metabolic and
Accepted: 17 October 2022
vasculature reactions, which in turn improve the functionality of neurons [3]. Therefore,
Published: 25 October 2022
the oxygen level in cerebral tissue is a crucial element that impacts nerve and glial cell
Publisher’s Note: MDPI stays neutral functions [2].
with regard to jurisdictional claims in Brain injury is a common cause of morbidity and mortality worldwide, especially in
published maps and institutional affil- the young population [4]. Secondary brain damage occurs in the hours, days or weeks after
iations. an event, and is associated with fatal outcomes [5]. Secondary insults may be mediated by
impaired cerebral metabolism, ischemia, intracranial hypertension and oxygen concentra-
tion disturbances such as hypoxia [6,7]. The combination of hypoxia and hypotension is
associated with enormously high mortality rates [8]. Recent data highlight the important
Copyright: © 2022 by the authors.
role of clinical protocols in improving oxygen delivery and resulting in lower mortality
Licensee MDPI, Basel, Switzerland.
in traumatic and nontraumatic brain-injured patients [9,10]. Clinical protocols guide the
This article is an open access article
rules for oxygen supplementation based on physiological processes such as increased
distributed under the terms and
conditions of the Creative Commons
oxygen supply (by monitoring of mean arterial pressure (MAP) and intracranial pressure
Attribution (CC BY) license (https://
(ICP), cerebral vasoreactivity and oxygen capacity) and reduction of oxygen demand (by
creativecommons.org/licenses/by/ pharmacological sedation and coma or hypothermia) [11]. Therefore, monitoring oxygen
4.0/). concentrations such as brain tissue oxygen (PbtO2 ) is an important aspect of brain injury

J. Pers. Med. 2022, 12, 1763. https://doi.org/10.3390/jpm12111763 https://www.mdpi.com/journal/jpm


J. Pers. Med. 2022, 12, 1763 2 of 20

clinical practice [12]. In addition, monitoring of mean arterial pressure and oxygenation of
both local and global tissues are essential for oxygenation and final outcomes [13].
The aim of this review is to discuss oxygen metabolism in the brain under
different conditions.

2. Oxygen Delivery and Autoregulation


The weight of the brain is only 2% of the human body, but cerebral tissue uses
25% of the glucose and about 20% of the oxygen delivered to function normally [14].
Oxygen consumption is 3.5 mL of oxygen/100 g tissue/1 min; therefore, the regulation
of blood flow and delivery of oxygen to cerebral tissue is crucial for brain function [15].
Importantly, 75–80% of the energy consumed by neurons is used at the synapses to restore
the neuronal membrane potentials lost during depolarization [16]. The continuous supply
of oxygen to the brain occurs via arterial blood and is transported to brain tissue by
diffusion. Diffusion is linked to the oxygen conductivity of cerebral tissue, determined
by the geometry of capillaries (distance and area) and the metabolism of tissue (oxygen
gradient from capillary to tissue) [17]. Extraction of oxygen is inversely proportional to
blood flow (when metabolism is constant) and directly proportional to metabolism (when
flow is constant) and the area between tissue and capillaries. Thus, a reduction in oxygen
delivery increases oxygen extraction. It should be noted that when cerebral blood flow
(CBF) is reduced by 50–60%, the consequent elevation of oxygen extraction is insufficient
to maintain proper cerebral oxygenation and a constant cerebral metabolic rate of oxygen
(CMRO2) [18]. Thus, cerebral oxygen delivery is determined by blood oxygen content and
cerebral blood flow. In physiological conditions, total blood flow in the brain is constant
because of the contribution of the large arteries to vascular resistance, as well as the impact
of the parenchymal arterioles on considerable basal tone.
Autoregulation of cerebral blood flow is the mechanism that enables the brain to
maintain relatively constant blood flow through changes in perfusion pressure [19]. In
a normotensive, physiological state, the ensuing cerebral perfusion pressure (CPP) is in
the range of 60 to 160 mmHg, and CBF is maintained at 50 mL per 100 g of brain tissue
per minute. Outside of this range, autoregulation is lost, and CBF starts to be dependent
on MAP in a linear mode [20]. A drop of CPP below the lower limit of 50 mmHg results
in cerebral ischemia [21]. This reduction of CBF is compensated for by elevated oxygen
extraction from the blood.
The individualization of care by targeting optimal, near to cerebral autoregulation
(CA)-guided CPP is connected with improved outcomes in TBI patients [22]. It is worth
remembering that combined brain tissue oxygen with ICP/CCP-guided therapy strongly
ameliorates favorable long-term outcomes [23]. In addition, in a recent meta-analysis, Xie
et al. documented that this combined therapy did not present any effects on mortality,
ICP/CPP and length of stay of patients after TBI [23].
Over a physiological range of partial oxygen pressure (PaO2 ) (75–100 mmHg;
7–13.33 kPa), PaO2 has little effect on global CBF as long as it does not fall below 50 mmHg
(6.67 kPa). This is because CBF is connected to the arterial content of oxygen rather than
PaO2 . The form of the hemoglobin–oxygen dissociation curve indicates that the arterial
content of oxygen is comparatively stable over the discussed PaO2 range [24].
The primary gradient determining the oxygen level in the brain may be enhanced by
a gradient-independent mechanism of cerebral vessel tone changes and increases in CBF
during functional neural activation (neurovascular coupling) [25,26]. The main role of this
mechanism is to transport higher levels of oxygen in advance of the elevated consumption
during neuronal activation [27].
Impairment of cerebral perfusion and metabolism following brain injury has been
documented repeatedly. Unfavorable outcomes after brain injury are connected with
hypoperfusion and decreased glucose metabolism and CMRO2 [28]. Recent data have
documented a connection between CMRO2 and Glasgow Coma Score (GCS) after traumatic
brain injury [29–32]. Soustiel et al. demonstrated that in TBI patients, CBF is somewhat
J. Pers. Med. 2022, 12, 1763 3 of 20

reduced during the first 24 h, and greater hypovolemia is observed following poor outcomes.
Importantly, a decrease in CMRO2 and the cerebral rate of glucose metabolism (CMRG)
correlates with worse outcomes [32].

3. Oxygen Consumption
Oxygen is transported to the cerebral cells by blood diffusion from the capillary to the
mitochondria, until it is consumed in the mitochondria as part of oxidative metabolism.
CMRO2 is the rate of consumption and energy homeostasis in the brain and in healthy,
awake people, averages 3.3 mL/100 g/min [33]. It is related to CBF. Under elevated
metabolic demand, the cerebral vasculature dilates to supply an appropriate increase
in CBF.
Importantly, with elevated neural activity, CMRO2 also rises [34,35]. In a normal,
unstimulated brain, energy is mostly provided by glucose oxidation. Nevertheless, the
metabolic rates of the oxygen-to-glucose ratio, CMRO2 /CMR(glc), called the oxygen-to-
glucose index (OGI), increase during activation and diverge from the textbook value of
6. In addition, the levels of lactates in the brain increase during sensory (e.g., visual)
stimulation [34]. This oxidative metabolism yields more energy as compared to glycolysis,
but precise measurements of this process are limited [36]. Mitochondria present a high
metabolic activity and a critical role in aerobic energy production, and their main function
is the production of adenosine triphosphate (ATP) through oxidative phosphorylation.
Mitochondrial dysfunction is a major factor in the occurrence of cell damage. Suc-
cessful resuscitation during ischemia/reperfusion demands the reestablishment of aerobic
metabolism by reperfusion of oxygenated blood. Mitochondria play a fundamental role as
effectors of reperfusion injury. Damage to the organelle impairs oxidative phosphorylation
and elimination of cytochrome c in the cytosol. The main mechanisms are oxidative stress
and Ca2+ overload [36].
Disturbances in oxygen delivery stop electron flow and interrupt the generation of the
“proton motive force” important in ATP production mentioned above. Of course, cells may
produce ATP anaerobically by glycolysis. However, this process is less effective, insufficient
for metabolic demands, and the final products are lactates.

4. Oxygen in the Cells


The role of mitochondria is to maintain maximal levels of ATP in the physiological
range of oxygen. It is important to remember that there are also other mechanisms responsi-
ble for oxygen consumption. Mostly, O2 is consumed by mitochondria, but 1–2% of oxygen
is incompletely reduced to superoxide anion (O2−) (Figure 1).

Figure 1. “Mitochondrial respiration” benefits when reducing nicotinamide adenine dinucleotide (NADH)
J. Pers. Med. 2022, 12, 1763 4 of 20

and flavin adenine dinucleotide (FADH2 ) components are created by the tricarboxylic acid (TCA)
cycle. In the inner mitochondrial membrane, electrons generated from NADH and FADH2 are
oxidized to NAD+ and FAD+ by complexes I and II. Afterward, these electrons are transferred
successively to complex III, cytochrome c and complex IV. Cytochrome c oxidase (COX, complex IV)
transmits electrons to molecular oxygen. This is an important enzyme in the mitochondrial electron
transport chain (ETC) connecting oxygen with oxidative phosphorylation [37]. The transmission of
electrons through the ETC is connected with proton transfer from the mitochondrial matrix, across
the inner membrane to the intermitochondrial membrane space. This translocation develops an
electrochemical gradient of protons (pH gradient and membrane potential). These molecules may drift
through the F1Fo-ATP synthase (complex V) or back to the mitochondrial matrix [38]. Importantly,
complex V connect protons transfer to the production of ATP from adenosine diphosphate (ADP)
and phosphate. Under normal oxygen levels, pyruvate, as a product of glycolysis, is transported
into the mitochondria, and is transformed into acetyl-CoA by the pyruvate dehydrogenase (PDH)
complex [39]. Afterward, acetyl-CoA connects with oxaloacetate and creates citrate—the first step in
the tricarboxylic acid cycle. Reducing equivalents in this cycle impacts ETC to production of ATP and
reactive oxygen species (ROS) for signaling, and the intermediates of TCA are used for biosynthetic
processes such as lipid synthesis [40].

Organisms develop important adaptation mechanisms [1]. One of these mechanisms


is metabolic suppression. Reduction of mitochondrial oxygen consumption in cells is
observed in oxygen levels between 1 and 3% in vitro. “Oxygen conformance” occurs
when the oxygen (<0.3%) level begins to limit the cytochrome c oxidase (COX) (complex
IV) [37]. Hypoxia, as a result of limited oxygen accessibility, results in reduction of oxidative
phosphorylation and loss of resynthesized phosphates, ATP and phosphocreatine. The
ATP-dependent Na/K pump is also changed and promotes the influx of Na, Ca and water
into cells, causing cytotoxic edema. In addition, ischemia impacts catabolism of adenine
nucleotides, resulting in the accumulation of hypoxanthine in cells. Cytosolic calcium
elevation promotes various pathways, such as activation of phospholipases and impor-
tantly the release of prostaglandins, lipases, proteases and endonucleases, which damages
structural elements of cells [41]. In addition, after increased expression of proinflammatory
gene products in the endothelium (leukocyte adhesion molecules, cytokines, endothelin
thromboxane A2) a proinflammatory state is observed. In contrast, prostacyclin and nitric
oxide are suppressed.

5. Hyperventilation/Hypoventilation
One of the most powerful factors affecting cerebral perfusion is hyperventilation/
hypoventilation, with an effect on CBF and PaCO2 . Hyperventilation is a common therapy
used to reduce elevated ICP or to relax a tense brain (hypocapnia-reduced CBF and CBV).
In traumatic brain injury (TBI) patients, hyperventilation generates a 34% decrease in CBF
and a 9% reduction in cerebral blood volume (CBV) when PaCO2 is decreased from 40 to
30 mmHg [42]. However, hyperventilation and hypocapnia, apart from vasoconstriction
and decreased CBF, also cause neuronal excitability and a longer duration of seizure eleva-
tion, an increase in excitatory amino acids and alkalosis of cerebrospinal fluid with a left
shift in the oxygen–hemoglobin dissociation curve (OHDC) [43,44]. All these mechanisms
may predispose to reduction in oxygen supply and delivery and a significant increase in
oxygen extraction.
It should be noted that carbon dioxide is a common molecule with a physiological
range of 35–45 mmHg. Hypocapnia (partial pressure of carbon dioxide <35 mmHg) and
mild hypercapnia (>45 mmHg) generate important nervous system disturbances. Recent
data have documented that hypercapnia presents neuroprotective mechanisms and may
improve CBF through cerebral vasodilatation. Hypercapnia also leads to brain edema,
elevated ICP, a right shift in the oxyhemoglobin dissociation curve, reduction of systemic
vascular resistance (SVR) and an increase in the tissue oxygen availability [45,46].
J. Pers. Med. 2022, 12, 1763 5 of 20

Hyperventilation is a double-edged sword with some short-term beneficial effects and


longer-term risks. The initial PaCO2 value in TBI in patients with normal ICP should be
within the normal range of 38–42 mmHg. Controlled hyperventilation during mechanical
ventilation in TBI patients (never below PaCO2 of 30 mmHg) is an approved therapeutic,
temporary (during the first 24 h after injury) life-saving intervention in severe intracranial
hypertension [46]. However, PaCO2 levels should be regulated and individualized in every
patient using multimodal neuromonitoring methods [11,47].

6. A Brief Search for “4-H” Factors Affecting CRMO2 and Cellular Oxygen Balance
Brain cells are especially susceptible to ischemic damage because of several unusual
features of their energy metabolism, high metabolic rate, restricted intrinsic energy stores
and critical relationship with the aerobic metabolism of glucose. Therefore, cell metabolism
and the consumption of crucial compounds can be altered by drugs or clinical status, which
can be summarized as “4-H”.

6.1. Hypoxia
The brain is one of the most sensitive organs to hypoxia, reoxygenation and oxidative
stress. As mentioned above, the brain has very high metabolic oxygen requirements, and it
is highly susceptible to hypoxic damage (Table 1).

Table 1. Potentially effect of disorders in oxygen delivery to the brain on selected pathways and
factors. Up arrows indicate the direction of the mechanism that may be intensified to varying degrees,
depending on the causative factor. The number of arrows defines the intensity of the processes.

Hypoxia Normoxia Hyperoxia


Oxidative stress ↑↑ ↑ ↑↑↑
Hypoxia-inducible factor
↑↑ ↑ ↑↑
(HIF)
Protein kinase B
↑↑ ↑ ↑↑
(Akt)
Extracellular signal-regulated
kinase ↑↑ ↑ ↑↑
(ERK)
Brain-derived neurotrophic
factor ↑↑ ↑ ↑↑
(BDNF)
Erythropoietin
↑↑ ↑ ↑↑
(Epo)
Neuroglobin
↑↑ ↑ ↑↑
(Ngb)
Nitric oxide (NO) ↑↑ ↑ ↑↑

Oxidative stress in mitochondria occurs in the state of redox imbalance and small
oxidant patterns such as superoxide radical, hydroxyl radical or nitric oxide radicals are
accumulated. It should be noted that both hypoxia and hyperoxia may induce oxida-
tive stress and apoptosis [48–50]. Acute hypoxia elevates ROS production in the brain,
and reoxygenation promotes this process. A low oxygen level leads to increased lipid
J. Pers. Med. 2022, 12, 1763 6 of 20

peroxidation, protein oxidation and nitric oxide levels and antioxidant defense systems.
Superoxide dismutase (SOD), reduced glutathione (GSH), glutathione peroxidase (GPx)
and reduced/oxidized glutathione (GSH/GSSG) ratio) are significantly inhibited in brain
cells. One of the crucial regulators of oxygen homeostasis and angiogenesis control in a
hypoxic state are HIFs (hypoxia-inducible factors). There are three transcription factors,
HIF-1, HIF-2 and HIF-3 [51]. These heterodimers are expressed by β subunits (HIF-1β,
HIF-2β and HIF-3β) and connected with α subunits, HIF-1α, HIF-2α and HIF-3α, directly
influencing hypoxia. The HIF-1 and HIF-2 are transcriptional regulators with unique target
genes. HIF-1 regulates the acute response to hypoxia (<24 h), and the network formatted
by HIF-1 predisposes to elevated perfusion and an increased oxygen level [52].
PaO2 has little effect on global CBF as long as it does not fall below 50 mmHg [53]. At
this point, there is a dramatic increase in blood flow with a further deterioration in PaO2 [53].
Reduction of ATP levels during hypoxia opens KATP channels on smooth muscle and causes
hypopolarization and vasodilatation [54]. Importantly, hypoxia further decreases PaO2 ,
and CBF may increase by up to 400% of the baseline level [55]. Changed CBF does not
affect metabolism, but hemoglobin saturation decreases from 100% (at PaO2 > 70 mmHg
(PaO2 > 9.33 kPa)) to 50% (at <50 mmHg (at <6.66 kPa)) [55]. In addition, the decrease
in PaO2 increases the production of local NO and adenosine. Chronic hypoxia increases
CBF by affecting capillary density [56,57]. Energy cell failure and delayed apoptosis are
connected with NO•, catalyzed by stimulation of nitric oxide synthase (nNOS) by lactic
acidosis and disruption of ionic transport [58,59].
In addition, neuronal membrane conversion leads to the release of glutamate, which
promotes activation of N-methyl-D-aspartate (NMDA) receptors and calcium influx pro-
moting lipases, proteases and endonucleases, precipitating free radical formation [59,60].
Finally, inflammation, critical mitochondrial dysfunction and ROS (superoxide, hydroxyl,
hydrogen peroxide and other) production with oxidation of lipids, proteins, cells and
deoxyribonucleic acid (DNA) are observed.
In animal models, oxidative stress parameters and the antioxidant system return to
the control system 24 h post brain injury [61]. Coimbra-Costa et al. documented that after
24 h of reoxygenation, oxidative stress is reduced, but apoptosis is preserved, especially
in the hippocampus [62]. The apoptotic rate in the hippocampus being higher than in the
cortex may be the reason for impairment of brain functions in hypoxic brain damage [63].
One of the crucial regulators of oxygen homeostasis and angiogenesis under a hypoxic
state are hypoxia-inducible factors (HIFs) [64]. The HIF-1 binds to hypoxia-responsive
elements (HRE) on gene promoters in the nucleus and promotes transcription of target
genes such as vascular endothelial growth factor (VEGF), glucose transporter 1 (GLUT1)
and others such as glycolysis enzymes, lactate dehydrogenase or erythropoietin [65–67].
HIF-1 is also crucial in glycolysis upregulation in astrocytes and Schwann cells [68]. In
contrast, HIF-2 and HIF-3 expressions start under chronic hypoxia in the endothelium.
Importantly, the switch from HIF-1 to HIF-2 and HIF-3 is observed during the adaptation
of the endothelium to prolonged hypoxia. HIF-1 covers the angiogenesis by formation of a
primary and very primitive network, and later expression of HIF-2 and HIF-3 stabilizes
and promotes maturation of this vasculature [69]. In addition, the network formatted by
HIF-1 predisposes to elevated perfusion and increased oxygen level [52].
Another mechanism under chronic hypoxia, which advances proteasomal degrada-
tion, is based on the carboxyl terminus of the Hsp70-interacting protein (Hsp70/CHIP
complex) [70]. The receptor for activated kinase C1 (RACK1) also leads to degradation
of HIF-1α and promotion of heat shock protein 90 (Hsp90), which secures the α sub-
unit [71,72]. Furthermore, RACK1 generates proteasomal degradation and ubiquitination
of HIF-1α [73]. Moreover, Kruppel-like factor 2 (KLF2), expressed in endothelial cells and
responsible for physiological vascularity formation, activates HIF-1 hypoxic degradation in
“a von Hippel–Lindau-independent, but proteasome-dependent manner” via interruption
of the connection Hsp90 with HIF-1 [74,75].
J. Pers. Med. 2022, 12, 1763 7 of 20

MicroRNA (miRNAs) is a family of noncoding RNA molecules with 18–22 nucleotides [76].
There is growing interest in the critical role of miRNA in the development and functioning
of the central nervous system as a gene regulator in “cleaving and silencing the gene expres-
sion” [77]. In contrast, atypical levels of miRNA are documented in various neurological
disorders [78]. The miRNA 210 is mainly expressed in a hypoxic state and is promoted by
HIF1 α and establishes a neuroprotective effect in hypoxia–ischemia damage [79,80].
The miRNA molecules decrease the apoptotic processes of neuronal cells with inhibi-
tion of caspases [81,82]. Therefore, with the growing interest in the association of miRNA
patterns with hypoxia/ischemia, these molecules may be clinical biomarkers for ischemia
and an individual miRNA therapeutics complex [83]. The protective effect of glucocorti-
coids (GCs) under hypoxia and ischemia/reperfusion has been shown recently [84]. GC
administration leads to increased tolerance to hypoxia in the central nervous system [85,86].
Acute hypoxia activates hypothalamic–pituitary–adrenal (HPA) with accumulation of up
to 24 h of corticosterone in serum [85]. Recent data have shown that hypoxic tolerance is
connected with upregulation of HIF-1 α and increased release of GC [85,87]. Direct crosstalk
between GC receptors and HIF-1 is potentially a basis of the biochemical pathways for GC
upregulation of HIF-1 target genes [88–90].
Clinical implications of hypoxia:
• Reduced brain tissue oxygenation is a predictor of poor outcome following severe
traumatic brain injury.
• Hypoxic–ischemic brain injury (HIBI) is associated with significant mortality and
morbidity [91].
• The LOCO2 study documented that targeting lower PaO2 improves outcomes in
patients with acute respiratory distress syndrome (ARDS) [92].
• The brain tissue oxygen tension (PbtO2 ) is crucial, the second monitored variable after
ICP, representing multimodality monitoring in TBI patients [11,93].
• Secondary hypoxia is connected with extended production of cytokines in CSF and
superior elevation of serum biomarkers such as myelin-basic protein (MBP) and
S100 [94].
• The MBP, S100 and neuron-specific enolase (NSE) biomarkers are more elevated in
patients with hypoxia and unfavorable outcomes (Extended Glasgow Outcome Coma
Score (GOSE) 1–4) [94]
• HIBI, as a two-hit model, is an effect of primary and secondary ischemic/hypoxic
damage predisposing to overall devastating severe injury of neurovascular units [91]
• Secondary brain hypoxia is connected with de novo neuronal and astroglial injury.
Importantly, secondary hypoxia is associated with cerebral proinflammatory response
but not parallel cerebral endothelial injury [91].
• Protocols based on PbtO2 and ICP monitoring significantly decrease cerebral hypoxia
time after TBI [95].
• Acute intermittent hypoxia (AIH) and task-specific training (TST) may synergistically
improve motor functions after central nervous system injury [96].

6.2. Hyperoxia
The concept of hyperoxia toxicity is defined by endogenous production of ROS [48,97].
Experimental examination of mitochondrial structure after 100% oxygen therapy
showed swollen and huge mitochondria and diluted and damaged mitochondria mem-
branes and cristae, which were directly connected with myelin, axonal and cellular or-
ganelle injury in the cortical brain [98]. Hyperoxia is connected with inhibition of Akt
expression and/or phosphorylation, the reverse of low oxygen levels [99,100]. Experi-
mental research has documented that in rat models of hyperoxia (FiO2 0.4–0.8), p-Akt
expression decreases steadily, over time until 12 h, then reverses to baseline value [100,101].
Thus, Akt signaling increases in hypoxia and is depressed in hyperoxia [102].
Mitogen-activated protein kinases (MAPKs) are involved in the PI3K-Akt signaling
pathway, an important pathway with a neuroprotective role against hypoxia or oxidative
J. Pers. Med. 2022, 12, 1763 8 of 20

stress [103]. Recent data on rat models showed that hyperoxia (FiO2 0.4–0.8) decreases the
p-ERK1/2 activation until 12 h and is followed by recovery in the subsequent 12 h [101].
Furthermore, hyperoxia in rat brain models impacts BDNF and neurotrophins 3 and
4 downregulation, and proceeds with correction in the subsequent 15–20 h, predispos-
ing to hyperoxia-linked apoptotic neurodegeneration [101,104]. Erythropoietin receptor
(EpoR) binding, observed in different brain areas, also plays an important role in oxygen
metabolism [105]. Under hypoxia, it is upregulated because HIF-1α binds to the Epo, show-
ing a neuroprotective effect contrary to ischemia hypoxia/reoxygenation injury [106–108].
Experimental data showed that hyperoxia upregulates Epo in mice treated with FiO2 0.5
for 3 weeks, elevating HIF-2α, but during 4 weeks of treatment with FiO2 = 0.3, only
EpoR expression increases [100]. Noteworthy is NO, which ameliorates oxygen delivery by
improving cerebral blood flow in the microvasculature [109]. In addition, NOS presents
a neuroprotective effect by improving vessel autoregulation [110]. It triggers different
mechanisms such as BDNF expression, HIF stabilization, S-nitrosylation of the HIF, block-
ing HIF-1α degradation, interaction with MAPK and phosphoinositide 3-kinase (PI3K)
signaling, and EpoR expression upregulation [111–116]. In a nonphysiological state pre-
senting hyperactivity of selected NOS, the NO starts to be neurotoxic as a free radical [109].
High oxygen concentration controls NOS expression and inhibits NO via surplus release
of superoxide anions inhibiting NO vasorelaxation and promoting vasoconstriction in the
brain [117,118]. Importantly, the connection of superoxide anions with NO promotes per-
oxynitrite (ONOO−) production with destructive properties [119–121]. In animal research,
NO is connected with hyperoxia-induced proliferation and proinflammatory responses in
astrocytes via cyclooxygenase-2 and prostaglandin E2 suppression [122].
Clinical implications of hyperoxia:
• Hyperoxia is associated with higher mortality and worse short-term functional out-
comes, especially in patients who receive uncontrolled oxygen delivery during the first
24 h after brain injury (probably because of hyperoxia-induced oxygen-free radical
toxicity with or without vasoconstriction) [123].
• Potential toxicity of a high oxygen concentration (patients receiving FiO2 of more
than 0.6).
• Previous studies documented that higher inspired oxygen concentration is associated
with acute lung injury, with mild to severe diffuse alveolar damage (DAD) [124].
• High oxygen levels within 72 h after aneurysmal rupture is an uninfluenced predictor
of cerebral vasospasm [125].
• In addition, liberal oxygen therapy increased 30-day mortality compared with conser-
vative therapy [126].
• Controversial high-dose oxygen therapy recommendations to reduce surgical site
infections (SSIs) by World Health Organization global guidelines for the prevention of
surgical site infection [127].
• Hyperoxemia may reduce cardiac output and increase systemic vascular resistance in
patients with cardiovascular failure [128].

6.3. Hyperthermia
Increased body temperature is frequently observed in patients following brain dam-
age due to direct hypothalamic injury, cerebral inflammation or secondary infection
indicating fever.
Systemic hyperthermia is common after brain damage. In patients with brain injury, it
is associated with poor neurological outcomes because it predisposes to worse secondary
damage [129] (Figure 2).
J. Pers. Med. 2022, 12, 1763 9 of 20

Figure 2. Hyperthermia is associated with poor neurological outcomes because it predisposes


to greater secondary damage. Temperature changes lead to elevation of cytokine release, higher
neutrophil activity and elevated metabolic expenditure. Hyperthermia also increases ROS generation
and apoptosis.

It should be noted that hyperthermia is not always connected with fever. Fever is an
adaptive reaction with, e.g., elevated neutrophil migration, activation of T-lymphocytes
and increased interleukin-1 and interferon production [130]. Temperature changes lead
to elevated cytokine release, higher neutrophil activity and elevated metabolic expendi-
ture, elevated white blood cell accumulation, increased vascular permeability, and axonal
damage [129]. Temperature changes also lead to cerebral blood flow conversion and hence
cause changes to cerebral oxygenation. Recent animal research has documented that
hyperthermia is associated with CD18 and intercellular adhesion molecule-1 (ICAM-1)
activation, as well as with an increase in ionized calcium-binding adapter protein-1 (IBA-1)
reactive microglia in the cortex [131]. Hyperthermia also increases ROS generation and
apoptosis, for example by c-Jun N-terminal kinase (JNK) activation [132,133]. Wettervik
et al. documented that hyperthermia leads to energy metabolism disturbances with no
associations with higher ICP and lower CPP [134]. Importantly, higher temperature was
connected with lower glucose concentration in cerebral tissue and a higher percentage of
the lactate-pyruvate ratio >25 after 5 days [134]. In addition, the authors did not show a
connection between hyperthermia and worse clinical outcomes.
The metabolic rate rises by around 20–25% during increased baseline core temperature
of over 1.5–2 ◦ C [135]. Recent data have shown that rising core temperature impacts
increased cerebral glucose utilization and CMRO2 by 5 to 10% per degree Celsius [136].
Nunneley et al. observed that a temperature elevated by more than 2 ◦ C is associated
with a higher glucose metabolic rate in the hypothalamus, thalamus, corpus callosum,
cingulate gyrus and cerebellum and lower in the caudate, putamen, insula and posterior
cingulum [137]. In addition, an increase in brain metabolism by 10% following a 2 ◦ C
higher temperature may be connected with an important reduction of blood flow to support
oxygenation [137]. Further, Spiotta et al. documented that hyperthermia did not reduce
brain tissue oxygen [138]. It should be noted that higher body temperature leads to a better
ability to maintain O2 uptake (VO2 ) because a higher fraction of the delivered O2 is extracted
J. Pers. Med. 2022, 12, 1763 10 of 20

before the beginning of O2 supply subjection [139]. Cardiovascular adjustments, as well as


sympathetic nerve activity during hyperthermia, also impact coupling between CMRO2 and
CBF. The activity of sympathetic nerves increases under hyperthermia. Adrenergic nerves
surround the vascular system, especially cerebral arteries [140]. Some authors suggest that
vasoconstriction under hyperthermia causes decreased CBF [141]. However, there are a
few doubts. First, in a hypermetabolic state under hyperthermia, different agents such as
histamine, nitric oxide or prostanoids may counteract vasoconstriction [142]. Second, blood
pressure significantly influences the cerebral vascular system. Third, the heterogenous
response of cerebral vascularity may be modified by hyperthermia and changes in the
density of alpha- and beta-adrenergic receptors [143]. Elevated body temperature impairs
blood–brain barrier (BBB) integrity, especially with dehydration [144,145]. Finally, Bein
et al. documented that the normalization of PaCO2 to eucapnia leads CBF to recuperate to
a physiological state [18].
Clinical implications of hyperthermia:
• Systemic complications such as fever frequently occur in the early phase after brain
damage and worsen secondary brain injury [134,146].
• Up to 50% of patients after acute brain injury experience fever during hospitalization [147].
• Brain temperature variations (>1 ◦ C) are associated with poor functional outcomes [148].
• In sum, higher body temperature is associated with elevated metabolic demand and
endogenous stress levels, blood pressure level changes, increases in cardiac output and
heart rate, hyperventilation, the synaptic release of excitatory amino acids, increased
ICP levels, ischemic cortical depolarizations, and BBB breakdown [146,149–154].
• Hyperthermia without oxygen delivery mismatch does not seem to induce significant
neurochemical alterations such as glucose, lactate, pyruvate and glutamate levels [151].
• PbtO2 may be an important element to be monitored during a high body temperature
episode to provide a view into oxygen metabolism in the brain [155].
• PbtO2 variations are observed under increased temperature increases in severe TBI
patients. PbtO2 may rise on average in every third and decrease in every sixth episode
of high temperature. Recent data have documented that the PbtO2 slope may occur
simultaneously with CPP and MAP reduction [156].
• Temperature management to prevent fever is crucial for patients with severe trau-
matic brain injury. The international guidelines for severe brain injury highlight the
importance of core temperature measurement and treatment above 38 ◦ C [11,46].

6.4. Hypothermia
The main objective of current international clinical guidelines is to ameliorate final
outcomes by inhibiting secondary injury, especially in the acute phase after damage. These
protocols also include correction of temperature and therapeutic hypothermia. Moderate
to deep hypothermia suppresses inflammation and decreases excitotoxicity and the pro-
duction of free radicals, which is one of the mechanisms of neuroprotection [157]. Different
levels of hypothermia improve neuronal tolerance to ischemia and inhibited neuronal
death [158,159]. Cerebral hypothermia decreases ICP, maintains BBB function and ame-
liorates glucose utilization [160–163]. In addition, lower temperature suppresses hypoxic
brain depolarization, releases neurotransmitters and decreases metabolism by protease
activation and a high energy phosphate depletion rate [164,165]. Authors have even noted
that deep hypothermia affects cerebral ATP production and improves survival after cardiac
arrest by three to four times [164]. Importantly, hypothermia during ischemia reduces
lipid peroxidation and essentially decreases ROS production [159,166]. Hypothermia re-
duces JNK activation and the apoptotic rate [132]. Hypothermia also activates a cascade
of neuroinflammation and may improve M1/M2 macrophage polarization to a favorable
phenotype [167].
Lower temperature improves cerebral metabolism after TBI and cerebral ischemia.
In animal models, the metabolic rate for glucose (CMRglc ) and CMRO2 is decreased, but
significantly, ATP distribution is decreased more than synthesis is [168]. Furthermore,
J. Pers. Med. 2022, 12, 1763 11 of 20

under a normoxic state, hypothermia decreases oxygen consumption in the brain as well as
collateral depletion of CBF and delivery of oxygen (elevated cerebrovascular resistance and
trace changes in oxygen extraction in the brain) [169]. Temperate hypoxia causes elevated
CBF and oxygen extraction, followed by reduced cerebrovascular resistance [170]. Chihara
et al., in an animal model of reduction in cerebral temperature by 1.6◦ ± 0.1◦ and hypoxia,
documented that hypothermia results in decreased oxygen delivery, oxygen consumption
and CBF. In addition, a significant improvement in cerebral vascular resistance is observed
as well as no oxygen extraction shift [171]. Recent data by Hashem et al., using near-
infrared spectroscopy (NIRS) and magnetic resonance imaging (MRI) methods, presented a
significant decrease of CMRO2 in the cortex of around 37 and 32% of hypothermic mice and
rats, respectively [172]. Therefore, targeting brain tissue oxygenation by different methods
such as an NIRS device may be an important aspect of brain damage treatment guidelines
for improving cerebral oxygenation, monitoring cerebrovascular reactivity (CVR) and final
outcomes [173,174].
Clinical implications of hypothermia
• Therapeutic hypothermia is a crucial component of current clinical practice guidelines.
• Therapeutic hypothermia uses different cooling methods to maintain brain tempera-
ture at target levels.
• Therapeutic hypothermia improves neurological outcomes [175]. In contrast, acciden-
tal hypothermia at admission after TBI results in higher hospital mortality [176].
• Recently published data do not promote early prophylactic hypothermia within the
first 6 h after damage in TBI patients [177].
• Body temperature of 35 to 35.5 ◦ C after TBI reduces intracranial hypertension and pre-
serves adequate CPP without cardiac dysfunction and oxygen debt [178]. In addition,
hypothermia reduces high ICP [177].
• Recent meta-analyses have documented the importance of temperature measurement
to avoid hypothermia in prehospital management [176].

7. Future Therapies
The oxygen-related mechanisms discussed have been a target for therapy in brain
injuries. One crucial element in the management of patients with various forms of cerebral
damage is the maintenance of oxygen homeostasis, supply and consumption, translating
into normal mitochondrial metabolism. Both hypoxia and hyperoxia may present a negative
effect on the final neurological outcome. Recent findings have shown the role of oxygen
therapy in neuroprotection, related to normobaric hyperoxia (NBHO). Patients with acute
brain injury treated with high oxygen levels (FiO2 0.6–1.0) for two hours presented with
improved redox balance and reduced lactate/pyruvate ratio (∆LPR −3.07 p = 0.015) [179].
The NBHO method is based on continuous administration of oxygen in normal atmospheric
pressure. Experimental data have documented the benefits of NBHO in ischemic stroke,
hemorrhagic strokes and brain trauma [180,181].
Yang et al. in an animal model experimentally documented the effect of normobaric
oxygen therapy (60%) on neurological functions, edema and HIF-1α, aquaporin 4 (AQP4)
and Na+/H+ exchanger 1 (NHE1) expression (p < 0.05, respectively). These authors showed
that therapy inhibits NHE1 expression and Na+ influx. These effects result in the reduction
of brain edema following the movement of water by AQP4 [180]. Hyperbaric oxygen
therapy (HBOT) is another therapy proposed in TBI. HBOT is 100% oxygen inhalation
under a pressure greater than 1 absolute atmosphere. HBOT suppresses inflammation and
defends BBB integrity and supports angiogenesis and neurogenesis [182,183]. Recent data
in an animal model documented that oxygen therapy at an early stage after brain damage
significantly decreased NF -κB and extracellular histones H1, H2A and H4 expression [184].
Histones are structural proteins in nuclei, an important factor in inflammation caused
by hypoxia and ischemia [185]. In addition, HBOT inhibits the apoptotic mechanisms in
neuronal cells and preserves the properties of mitochondrial membranes, reducing sec-
ondary damage [186,187]. Of course, the clinical effectiveness of HBOT is still controversial.
J. Pers. Med. 2022, 12, 1763 12 of 20

Rockswold et al. documented in a small study that HBOT did not improve outcomes in a
group of patients with closed head injury [188]. However, in other phase II clinical trials in
2013, Roackswold et al. demonstrated that combined HBOT with normobaric hyperoxia
(NBHT) therapy improves oxidative metabolism and oxygen brain tissue partial pressure
levels [189]. In addition, this therapy decreased intracranial hypertension, mortality and
improved outcomes (measured by GOSE) [189]. Another study showed that HBOT sig-
nificantly improved post-traumatic stress disorder symptoms, cognitive functions and
decreased depression and anxiety [190].
The controversial effects of HBOT may be explained by the hyperoxic–hypoxic paradox
(HHP). Recent research has shown that repetitive and periodic hyperoxia may induce
molecular mechanisms and activate mediators similarly to hypoxia [191]. Activation of
HIF, VEGF, SIRT, mitochondrial biogenesis and stem cell proliferation is observed during
intermittent hyperoxia.
Another therapeutic option is the significant role of lactate in cerebral energy
metabolism [192]. Experimental lactate supplementation in ischemic brain damage impacts
decreased glutamate- and gamma-aminobutyric acid (GABA) release with improvement
in electroencephalogram (EEG) [193]. Furthermore, Berthet et al. documented that lactate
supplementation inhibits neuronal death in oxygen and glucose delivery disturbances [194].
The same treatment in middle cerebral artery occlusion and ischemia models also presents
a significant neuroprotective effect [195,196]. Ichai et al., in randomized controlled trials,
presented that hypertonic sodium lactate (HSL) treatment is more potent in reducing ele-
vated ICP than mannitol in a group of TBI patients [197]. In addition, this effect lasts longer
and is connected with improvement in jugular venous O2 saturation, glucose and lactate
levels in plasma and pH. Patients also presented better neurological final outcomes [197].
The infusion of HSL for 3 h impacts extracellular metabolites. One theory is that these
solutions contain metabolizable lactate and Na ions. Lactate in the brain induces an imbal-
ance between anions and positive charges and counteracts the harmful cellular swelling by
compensation of anion efflux [197] (Supplementary Materials).
Recent data have shown elevated lactate, pyruvate and glucose levels in the brain with
associated lower glutamate and PbtO2 values as well as ICP. Bouzat et al. documented that
these effects may be the result of a brain metabolism shift to elevated lactate utilization,
sparing the effect of glucose. In addition, the inhibition of cerebral oxygenation may be
secondary to alkalosis, which increases the affinity of oxygen to hemoglobin and suggests a
beneficial effect [198]. In summary, hypertonic sodium lactate infusion reduces glutamate-
related excitotoxicity, improves cerebral perfusion, buffers metabolic acidosis, decreases
cerebral edema and ICP and improves cardiac performance [199–201].

8. Conclusions
Oxygen is crucial for the functionality of cerebral cells. Therefore, the mechanisms
leading to disruption of oxygen supply and consumption are the subject of continuous
intensive research. There is a growing need for novel therapeutic methods to reduce the
cascade of pathological cellular processes.

Supplementary Materials: The following supporting information can be downloaded at: https:
//www.mdpi.com/article/10.3390/jpm12111763/s1, Table S1: Novel therapy approaches.
Author Contributions: Conceptualization, C.R. and D.S.-G.; methodology, D.S.-G. and C.R.; valida-
tion, C.R., W.D. and D.S.-G.; formal analysis, D.S.-G.; writing—original draft preparation, D.S.-G.;
writing—review and editing, C.R., D.S.-G., W.D., J.G. and R.B.; visualization, D.S.-G., W.D., C.R. and
J.G.; supervision, D.S.-G., C.R., W.D. and R.B.; project administration, D.S.-G. All authors have read
and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
J. Pers. Med. 2022, 12, 1763 13 of 20

Data Availability Statement: Not applicable.


Conflicts of Interest: The authors declare no conflict of interest.

References
1. Semenza, G.L. Life with Oxygen. Science 2007, 318, 62–64. [CrossRef] [PubMed]
2. Özugur, S.; Kunz, L.; Straka, H. Relationship between oxygen consumption and neuronal activity in a defined neural circuit.
BMC Biol. 2020, 18, 76. [CrossRef] [PubMed]
3. Roy, C.S.; Sherrington, C.S. On the Regulation of the Blood-supply of the Brain. J. Physiol. 1890, 11, 85–158. [CrossRef]
4. Maas, A.I.R.; Menon, D.K.; Adelson, P.D.; Andelic, N.; Bell, M.J.; Belli, A.; Bragge, P.; Brazinova, A.; Büki, A.; Chesnut, R.M.; et al.
Traumatic brain injury: Integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017, 16, 987–1048.
[CrossRef]
5. Chesnut, R.M.; Marshall, L.F.; Klauber, M.R.; Blunt, B.A.; Baldwin, N.; Eisenberg, H.M.; Jane, J.A.; Marmarou, A.; Foulkes, M.A.
The Role of Secondary Brain Injury in Determining Outcome from Severe Head Injury. J. Trauma Inj. Infect. Crit. Care 1993,
34, 216–222. [CrossRef] [PubMed]
6. Volpi, P.C.; Robba, C.; Rota, M.; Vargiolu, A.; Citerio, G. Trajectories of early secondary insults correlate to outcomes of traumatic
brain injury: Results from a large, single centre, observational study. BMC Emerg. Med. 2018, 18, 52. [CrossRef]
7. Maloney-Wilensky, E.; Gracias, V.; Itkin, A.; Hoffman, K.; Bloom, S.; Yang, W.; Christian, S.; Leroux, P.D. Brain tissue oxygen and
outcome after severe traumatic brain injury: A systematic review. Crit. Care Med. 2009, 37, 2057–2063. [CrossRef]
8. Jeremitsky, E.; Omert, L.; Dunham, C.M.; Protetch, J.; Rodriguez, A. Harbingers of Poor Outcome the Day after Severe Brain
Injury: Hypothermia, Hypoxia, and Hypoperfusion. J. Trauma Inj. Infect. Crit. Care 2003, 54, 312–319. [CrossRef]
9. Bogossian, E.G.; Diaferia, D.; Djangang, N.N.; Menozzi, M.; Vincent, J.-L.; Talamonti, M.; Dewitte, O.; Peluso, L.; Barrit, S.; Al
Barajraji, M.; et al. Brain tissue oxygenation guided therapy and outcome in non-traumatic subarachnoid hemorrhage. Sci. Rep.
2021, 11, 16235. [CrossRef]
10. Martini, R.P.; Deem, S.; Treggiari, M.M. Targeting Brain Tissue Oxygenation in Traumatic Brain Injury. Respir. Care 2012,
58, 162–172. [CrossRef]
11. Chesnut, R.; Aguilera, S.; Buki, A.; Bulger, E.; Citerio, G.; Cooper, D.J.; Arrastia, R.D.; Diringer, M.; Figaji, A.; Gao, G.; et al. A
management algorithm for adult patients with both brain oxygen and intracranial pressure monitoring: The Seattle International
Severe Traumatic Brain Injury Consensus Conference (SIBICC). Intensiv. Care Med. 2020, 46, 919–929. [CrossRef]
12. Rakhit, S.; Nordness, M.F.; Lombardo, S.R.; Cook, M.; Smith, L.; Patel, M.B. Management and Challenges of Severe Traumatic
Brain Injury. Semin. Respir. Crit. Care Med. 2021, 42, 127–144. [CrossRef]
13. Sekhon, M.S.; Gooderham, P.; Menon, D.K.; Brasher, P.M.A.; Foster, D.; Cardim, D.; Czosnyka, M.; Smielewski, P.; Gupta, A.K.;
Ainslie, P.N.; et al. The Burden of Brain Hypoxia and Optimal Mean Arterial Pressure in Patients With Hypoxic Ischemic Brain
Injury After Cardiac Arrest. Crit. Care Med. 2019, 47, 960–969. [CrossRef]
14. Hyder, F.; Rothman, D.L.; Bennett, M.R. Cortical energy demands of signaling and nonsignaling components in brain are
conserved across mammalian species and activity levels. Proc. Natl. Acad. Sci. USA 2013, 110, 3549–3554. [CrossRef]
15. Rink, C.; Khanna, S. Significance of Brain Tissue Oxygenation and the Arachidonic Acid Cascade in Stroke. Antioxid. Redox Signal
2011, 14, 1889–1903. [CrossRef]
16. Harris, J.J.; Jolivet, R.; Attwell, D. Synaptic Energy Use and Supply. Neuron 2012, 75, 762–777. [CrossRef]
17. Gjedde, A. The pathways of oxygen in brain. I. Delivery and metabolism of oxygen. In Advances in Experimental Medicine and
Biology; Springer: Boston, MA, USA, 2005; p. 566.
18. Bain, A.R.; Morrison, S.; Ainslie, P.N. Cerebral oxygenation and hyperthermia. Front. Physiol. 2014, 5, 92. [CrossRef]
19. Paulson, O.B.; Strandgaard, S.; Edvinsson, L. Cerebral autoregulation. Cerebrovasc. Brain Metab. Rev. 1990, 2, 161–192.
20. Cipolla, M.J.; Osol, G. Vascular Smooth Muscle Actin Cytoskeleton in Cerebral Artery Forced Dilatation. Stroke 1998, 29, 1223–1228.
[CrossRef]
21. Baron, J.-C. Perfusion Thresholds in Human Cerebral Ischemia: Historical Perspective and Therapeutic Implications. Cerebrovasc.
Dis. 2001, 11 (Suppl. 1), 2–8. [CrossRef]
22. Tas, J.; Beqiri, E.; van Kaam, R.C.; Czosnyka, M.; Donnelly, J.; Haeren, R.H.; van der Horst, I.C.; Hutchinson, P.J.; van Kuijk, S.M.;
Liberti, A.L.; et al. Targeting Autoregulation-Guided Cerebral Perfusion Pressure after Traumatic Brain Injury (COGiTATE): A
Feasibility Randomized Controlled Clinical Trial. J. Neurotrauma 2021, 38, 2790–2800. [CrossRef] [PubMed]
23. Xie, Q.; Wu, H.-B.; Yan, Y.-F.; Liu, M.; Wang, E.-S. Mortality and Outcome Comparison Between Brain Tissue Oxygen Combined
with Intracranial Pressure/Cerebral Perfusion Pressure–Guided Therapy and Intracranial Pressure/Cerebral Perfusion Pressure–
Guided Therapy in Traumatic Brain Injury: A Meta-Analysis. World Neurosurg. 2017, 100, 118–127. [CrossRef] [PubMed]
24. McDowall, D.G. Interrelationships between blood oxygen tensions and cerebral blood flow. Oxyg. Meas. Blood Tissues 1966,
4, 205–219.
25. Attwell, D.; Buchan, A.M.; Charpak, S.; Lauritzen, M.J.; MacVicar, B.A.; Newman, E.A. Glial and neuronal control of brain blood
flow. Nature 2010, 468, 232–243. [CrossRef] [PubMed]
26. Muoio, V.; Persson, P.B.; Sendeski, M.M. The neurovascular unit—concept review. Acta Physiol. 2014, 210, 790–798. [CrossRef]
27. Sokoloff, L. Energetics of Functional Activation in Neural Tissues. Neurochem. Res. 1999, 24, 321–329. [CrossRef]
J. Pers. Med. 2022, 12, 1763 14 of 20

28. Hattori, N.; Huang, S.-C.; Wu, H.-M.; Yeh, E.; Glenn, T.C.; Vespa, P.M.; McArthur, D.; E Phelps, M.; A Hovda, D.; Bergsneider,
M. Correlation of regional metabolic rates of glucose with glasgow coma scale after traumatic brain injury. J. Nucl. Med. 2003,
44, 1709.
29. Bergsneider, M.; Hovda, D.A.; Lee, S.M.; Kelly, D.F.; McArthur, D.; Vespa, P.M.; Lee, J.H.; Huang, S.-C.; Martin, N.; Phelps, M.E.;
et al. Dissociation of Cerebral Glucose Metabolism and Level of Consciousness During the Period of Metabolic Depression
Following Human Traumatic Brain Injury. J. Neurotrauma 2000, 17, 389–401. [CrossRef]
30. Bergsneider, M.; Hovda, D.A.; Shalmon, E.; Kelly, D.F.; Vespa, P.M.; Martin, N.A.; Phelps, M.E.; McArthur, D.L.; Caron, M.J.;
Kraus, J.F.; et al. Cerebral hyperglycolysis following severe traumatic brain injury in humans: A positron emission tomography
study. J. Neurosurg. 1997, 86, 241–251. [CrossRef]
31. Obrist, W.D.; Langfitt, T.W.; Jaggi, J.L.; Cruz, J.; Gennarelli, T.A. Cerebral blood flow and metabolism in comatose patients with
acute head injury: Relationship to intracranial hypertension. J. Neurosurg. 1984, 61, 241–253. [CrossRef]
32. Soustiel, J.F.; Glenn, T.C.; Shik, V.; Boscardin, J.; Mahamid, E.; Zaaroor, M. Monitoring of Cerebral Blood Flow and Metabolism in
Traumatic Brain Injury. J. Neurotrauma 2005, 22, 955–965. [CrossRef]
33. Cold, G.E. Cerebral Metabolic Rate of Oxygen (CMRO2) in the Acute Phase of Brain Injury. Acta Anaesthesiol. Scand. 1978,
22, 249–256. [CrossRef]
34. Shulman, R.G.; Hyder, F.; Rothman, D.L. Lactate efflux and the neuroenergetic basis of brain function. NMR Biomed. 2001,
14, 389–396. [CrossRef]
35. Thompson Jeffrey, K.; Peterson Matthew, R.; Freeman Ralph, D. Single-Neuron Activity and Tissue Oxygenation in the Cerebral
Cortex. Science 2003, 299, 1070–1072. [CrossRef]
36. Hyder, F.; Kida, I.; Behar, K.L.; Kennan, R.P.; Maciejewski, P.K.; Rothman, D.L. Quantitative functional imaging of the brain:
Towards mapping neuronal activity by BOLD fMRI. NMR Biomed. 2001, 14, 413–431. [CrossRef]
37. Hochachka, P.W.; Buck, L.T.; Doll, C.J.; Land, S.C. Unifying theory of hypoxia tolerance: Molecular/metabolic defense and rescue
mechanisms for surviving oxygen lack. Proc. Natl. Acad. Sci. USA 1996, 93, 9493–9498. [CrossRef]
38. Boyer, P.D. The Atp Synthase—A Splendid Molecular Machine. Annu. Rev. Biochem. 1997, 66, 717–749. [CrossRef]
39. Wu, I.C.; Ohsawa, I.; Fuku, N.; Tanaka, M. Metabolic analysis of 13C-labeled pyruvate for noninvasive assessment of mitochondrial
function. Ann. N. Y. Acad. Sci. 2010, 1201, 111–120. [CrossRef]
40. Wellen, K.E.; Thompson, C.B. Cellular metabolic stress: Considering how cells respond to nutrient excess. Mol. Cell 2010,
40, 323–332. [CrossRef]
41. Sas, K.; Robotka, H.; Toldi, J.; Vécsei, L. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with
focus on neurodegenerative disorders. J. Neurol. Sci. 2007, 257, 221–239. [CrossRef]
42. Diringer, M.N.; Yundt, K.; Videen, T.O.; Adams, R.E.; Zazulia, A.R.; Deibert, E.; Aiyagari, V.; Dacey, R.G.; Grubb, R.L.; Powers,
W.J. No reduction in cerebral metabolism as a result of early moderate hyperventilation following severe traumatic brain injury. J.
Neurosurg. 2000, 92, 7–13. [CrossRef] [PubMed]
43. Zhang, Z.; Guo, Q.; Wang, E. Hyperventilation in neurological patients: From physiology to outcome evidence. Curr. Opin.
Anesthesiol. 2019, 32, 568–573. [CrossRef] [PubMed]
44. Mas, A.; Saura, P.; Joseph, D.; Blanch, L.; Baigorri, F.; Artigas, A.; Fernandez, R. Effect of acute moderate changes in PaCO2 on
global hemodynamics and gastric perfusion. Crit. Care Med. 2000, 28, 360–365. [CrossRef] [PubMed]
45. Howarth, C.; Sutherland, B.A.; Choi, H.B.; Martin, C.; Lind, B.L.; Khennouf, L.; LeDue, J.M.; Pakan, J.M.; Ko, R.W.; Ellis-Davies, G.;
et al. A Critical Role for Astrocytes in Hypercapnic Vasodilation in Brain. J. Neurosci. 2017, 37, 2403–2414. [CrossRef] [PubMed]
46. Carney, N.; Totten, A.M.; O’Reilly, C.; Ullman, J.S.; Hawryluk, G.W.; Bell, M.J.; Bratton, S.L.; Chesnut, R.; Harris, O.A.; Kissoon, N.;
et al. Guidelines for the Management of Severe Traumatic Brain Injury, Fourth Edition. Neurosurgery 2017, 80, 6–15. [CrossRef]
47. Oddo, M.; Bösel, J. Monitoring of brain and systemic oxygenation in neurocritical care patients. Neurocritical Care 2014, 21, 103–120.
[CrossRef]
48. Chen, R.; Lai, U.H.; Zhu, L.; Singh, A.; Ahmed, M.; Forsyth, N.R. Reactive Oxygen Species Formation in the Brain at Different
Oxygen Levels: The Role of Hypoxia Inducible Factors. Front. Cell Dev. Biol. 2018, 6, 132. [CrossRef]
49. Banasiak, K.J.; Xia, Y.; Haddad, G.G. Mechanisms underlying hypoxia-induced neuronal apoptosis. Prog. Neurobiol. 2000,
62, 215–249. [CrossRef]
50. Chang, E.; Hornick, K.; Fritz, K.I.; Mishra, O.P.; Delivoria-Papadopoulos, M. Effect of Hyperoxia on Cortical Neuronal Nuclear
Function and Programmed Cell Death Mechanisms. Neurochem. Res. 2007, 32, 1142–1149. [CrossRef]
51. Serocki, M.; Bartoszewska, S.; Janaszak-Jasiecka, A.; Ochocka, R.J.; Collawn, J.F.; Bartoszewski, R. miRNAs regulate the HIF
switch during hypoxia: A novel therapeutic target. Angiogenesis 2018, 21, 183–202. [CrossRef]
52. Koh, M.Y.; Lemos, R.; Liu, X.; Powis, G. The hypoxia-associated factor switches cells from HIF-1α-to HIF-2α-dependent signaling
promoting stem cell characteristics, aggressive tumor growth and invasion. Cancer Res. 2011, 71, 4015–4027. [CrossRef]
53. Masamoto, K.; Tanishita, K. Oxygen Transport in Brain Tissue. J. Biomech. Eng. 2009, 131, 074002. [CrossRef]
54. Taguchi, H.; Heistad, D.D.; Kitazono, T.; Faraci, F.M. ATP-sensitive K+ channels mediate dilatation of cerebral arterioles during
hypoxia. Circ. Res. 1994, 74, 1005–1008. [CrossRef]
55. Johnston, A.J.; Steiner, L.A.; Gupta, A.K.; Menon, D.K. Cerebral oxygen vasoreactivity and cerebral tissue oxygen reactivity. Br. J.
Anaesth. 2003, 90, 774–786. [CrossRef]
56. Xu, K.; LaManna, J.C. Chronic hypoxia and the cerebral circulation. J. Appl. Physiol. 2006, 100, 725–730. [CrossRef]
J. Pers. Med. 2022, 12, 1763 15 of 20

57. Boero, J.A.; Ascher, J.; Arregui, A.; Rovainen, C.; Woolsey, T.A. Increased brain capillaries in chronic hypoxia. J. Appl. Physiol.
1999, 86, 1211–1219. [CrossRef]
58. Torres-Cuevas, I.; Parra-Llorca, A.; Sánchez-Illana, Á.; Nuñez-Ramiro, A.; Kuligowski, J.; Cháfer-Pericás, C.; Cernada, M.; Escobar,
J.; Vento, M. Oxygen and oxidative stress in the perinatal period. Redox Biol. 2017, 12, 674–681. [CrossRef]
59. Rocha-Ferreira, E.; Hristova, M. Plasticity in the Neonatal Brain following Hypoxic-Ischaemic Injury. Neural Plast. 2016,
2016, 4901014. [CrossRef]
60. Sekhon, M.S.; Ainslie, P.N.; Griesdale, D.E. Clinical pathophysiology of hypoxic ischemic brain injury after cardiac arrest: A
“two-hit” model. Crit. Care 2017, 21, 90. [CrossRef]
61. Chen, H.; Yoshioka, H.; Kim, G.S.; Jung, J.E.; Okami, N.; Sakata, H.; Maier, C.M.; Narasimhan, P.; Goeders, C.E.; Chan, P.H.
Oxidative Stress in Ischemic Brain Damage: Mechanisms of Cell Death and Potential Molecular Targets for Neuroprotection.
Antioxid. Redox Signal. 2011, 14, 1505–1517. [CrossRef]
62. Coimbra-Costa, D.; Alva, N.; Duran, M.; Carbonell, T.; Rama, R. Oxidative stress and apoptosis after acute respiratory hypoxia
and reoxygenation in rat brain. Redox Biol. 2017, 12, 216–225. [CrossRef] [PubMed]
63. Peña, F.; Ramirez, J.-M. Hypoxia-induced changes in neuronal network properties. Mol. Neurobiol. 2005, 32, 251–283. [CrossRef]
64. Majmundar, A.J.; Wong, W.J.; Simon, M.C. Hypoxia-inducible factors and the response to hypoxic stress. Mol. Cell 2010,
40, 294–309. [CrossRef] [PubMed]
65. Dengler, V.L.; Galbraith, M.D.; Espinosa, J.M. Transcriptional regulation by hypoxia inducible factors. Crit. Rev. Biochem. Mol.
Biol. 2014, 49, 1–15. [CrossRef] [PubMed]
66. Chávez, J.C.; Agani, F.; Pichiule, P.; LaManna, J.C. Expression of hypoxia-inducible factor-1α in the brain of rats during chronic
hypoxia. J. Appl. Physiol. 2000, 89, 1937–1942. [CrossRef] [PubMed]
67. Lukyanova, L.D.; Kirova, Y.I. Mitochondria-controlled signaling mechanisms of brain protection in hypoxia. Front. Neurosci. 2015,
9, 320. [CrossRef]
68. Brix, B.; Mesters, J.R.; Pellerin, L.; Jöhren, O. Endothelial cell-derived nitric oxide enhances aerobic glycolysis in astrocytes via
HIF-1α-mediated target gene activation. J. Neurosci. 2012, 32, 9727–9735. [CrossRef]
69. Koh, M.Y.; Powis, G. Passing the baton: The HIF switch. Trends Biochem. Sci. 2012, 37, 364–372. [CrossRef]
70. Luo, W.; Zhong, J.; Chang, R.; Hu, H.; Pandey, A.; Semenza, G.L. Hsp70 and CHIP Selectively Mediate Ubiquitination and
Degradation of Hypoxia-inducible Factor (HIF)-1α but Not HIF-2α 2. J. Biol. Chem. 2010, 285, 3651–3663. [CrossRef]
71. Liu, Y.V.; Semenza, G.L. RACK1 vs. HSP90: Competition for HIF-1α degradation vs. stabilization. Cell Cycle 2007, 6, 656–659.
[CrossRef]
72. Liu, Y.V.; Baek, J.H.; Zhang, H.; Diez, R.; Cole, R.N.; Semenza, G.L. RACK1 competes with HSP90 for binding to HIF-1α and is
required for O2-independent and HSP90 inhibitor-induced degradation of HIF-1α. Mol. Cell 2007, 25, 207–217. [CrossRef]
73. Ravi, R.; Mookerjee, B.; Bhujwalla, Z.M.; Sutter, C.H.; Artemov, D.; Zeng, Q.; Dillehay, L.E.; Madan, A.; Semenza, G.L.; Bedi,
A. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1α. Genes Dev. 2000, 14, 34–44.
[CrossRef]
74. Bhattacharya, R.; SenBanerjee, S.; Lin, Z.; Mir, S.; Hamik, A.; Wang, P.; Mukherjee, P.; Mukhopadhyay, D.; Jain, M.K. Inhibition of
Vascular Permeability Factor/Vascular Endothelial Growth Factor-mediated Angiogenesis by the Kruppel-like Factor KLF2. J.
Biol. Chem. 2005, 280, 28848–28851. [CrossRef]
75. Kawanami, D.; Mahabeleshwar, G.H.; Lin, Z.; Atkins, G.B.; Hamik, A.; Haldar, S.M.; Maemura, K.; LaManna, J.C.; Jain, M.K.
Kruppel-like Factor 2 Inhibits Hypoxia-inducible Factor 1α Expression and Function in the Endothelium. J. Biol. Chem. 2009, 284,
20522–20530. [CrossRef]
76. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The nuclear RNase III Drosha
initiates microRNA processing. Nature 2003, 425, 415–419. [CrossRef]
77. Davis, G.M.; Haas, M.A.; Pocock, R. MicroRNAs: Not “fine-tuners” but key regulators of neuronal development and function.
Front. Neurol. 2015, 6, 245. [CrossRef]
78. Xiao, S.; Ma, Y.; Zhu, H.; Sun, H.; Yin, Y.; Feng, G. miRNA functional synergistic network analysis of mice with ischemic stroke.
Neurol. Sci. 2015, 36, 143–148. [CrossRef]
79. Chan, S.Y.; Loscalzo, J. MicroRNA-210: A unique and pleiotropic hypoxamir. Cell Cycle 2010, 9, 1072–1083. [CrossRef]
80. Yang, G.-Y.; Zeng, L.; Liu, J.; Wang, Y.; Wang, L.; Weng, S.; Tang, Y.; Zheng, C.; Cheng, Q.; Chen, S. MicroRNA-210 as a novel
blood biomarker in acute cerebral ischemia. Front. Biosci. 2011, E3, 1265–1272. [CrossRef]
81. Qiu, J.; Zhou, X.-Y.; Zhou, X.-G.; Cheng, R.; Liu, H.-Y.; Li, Y. Neuroprotective effects of microRNA-210 on hypoxic-ischemic
encephalopathy. BioMed Res. Int. 2013, 2013, 350419. [CrossRef]
82. Tan, K.S.; Armugam, A.; Sepramaniam, S.; Lim, K.Y.; Setyowati, K.D.; Wang, C.W.; Jeyaseelan, K. Expression Profile of MicroRNAs
in Young Stroke Patients. PLoS ONE 2009, 4, e7689. [CrossRef]
83. Minhas, G.; Mathur, D.; Ragavendrasamy, B.; Sharma, N.K.; Paanu, V.; Anand, A. Hypoxia in CNS Pathologies: Emerging Role of
miRNA-Based Neurotherapeutics and Yoga Based Alternative Therapies. Front. Neurosci. 2017, 11, 386. [CrossRef] [PubMed]
84. Bobryshev, P.; Bagaeva, T.; Filaretova, L. Ischemic preconditioning attenuates gastric ischemia-reperfusion injury through
involvement of glucocorticoids. J. Physiol. Pharmacol. 2009, 60 (Suppl. 7), 155–160.
85. Rybnikova, E.A.; Mironova, V.I.; Pivina, S.G.; Ordyan, N.E.; Tulkova, E.I.; Samoilov, M.O. Hormonal Mechanisms of Neuroprotective
Effects of the Mild Hypoxic Preconditioning in Rats, 1st ed.; Springer Nature BV: Berlin/Heidelberg, Germany, 2008; p. 239.
J. Pers. Med. 2022, 12, 1763 16 of 20

86. Davis, C.; Hackett, P. Advances in the prevention and treatment of high altitude illness. Emerg. Med. Clin. 2017, 35, 241–260.
[CrossRef] [PubMed]
87. Rybnikova, E.; Mironova, V.; Pivina, S.; Tulkova, E.; Ordyan, N.; Nalivaeva, N.; Turner, A.; Samoilov, M. Involvement of the
hypothalamic-pituitary-adrenal axis in the antidepressant-like effects of mild hypoxic preconditioning in rats. Psychoneuroen-
docrinology 2007, 32, 813–823. [CrossRef] [PubMed]
88. Vanderhaeghen, T.; Beyaert, R.; Libert, C. Bidirectional Crosstalk Between Hypoxia Inducible Factors and Glucocorticoid
Signalling in Health and Disease. Front. Immunol. 2021, 12, 684085. [CrossRef]
89. Kodama, T.; Shimizu, N.; Yoshikawa, N.; Makino, Y.; Ouchida, R.; Okamoto, K.; Hisada, T.; Nakamura, H.; Morimoto, C.;
Tanaka, H. Role of the Glucocorticoid Receptor for Regulation of Hypoxia-dependent Gene Expression. J. Biol. Chem. 2003,
278, 33384–33391. [CrossRef]
90. Lim, W.; Park, C.; Shim, M.K.; Lee, Y.H.; Lee, Y.M.; Lee, Y. Glucocorticoids suppress hypoxia-induced COX-2 and hypoxia
inducible factor-1α expression through the induction of glucocorticoid-induced leucine zipper. Br. J. Pharmacol. 2014, 171, 735–745.
[CrossRef]
91. Hoiland, R.L.; Ainslie, P.N.; Wellington, C.L.; Cooper, J.; Stukas, S.; Thiara, S.; Foster, D.; Fergusson, N.A.; Conway, E.M.; Menon,
D.K.; et al. Brain Hypoxia Is Associated With Neuroglial Injury in Humans Post–Cardiac Arrest. Circ. Res. 2021, 129, 583–597.
[CrossRef]
92. Barrot, L.; Asfar, P.; Mauny, F.; Winiszewski, H.; Montini, F.; Badie, J.; Quenot, J.-P.; Pili-Floury, S.; Bouhemad, B.; Louis, G.;
et al. Liberal or Conservative Oxygen Therapy for Acute Respiratory Distress Syndrome. N. Engl. J. Med. 2020, 382, 999–1008.
[CrossRef]
93. Bardt, T.F.; Unterberg, A.W.; Härtl, R.; Kiening, K.L.; Schneider, G.H.; Lanksch, W.R. Monitoring of brain tissue PO2 in traumatic
brain injury: Effect of cerebral hypoxia on outcome. In Intracranial Pressure and Neuromonitoring in Brain Injury; Marmarou, A.,
Bullock, R., Avezaat, C., Baethmann, A., Becker, D., Brock, M., Hoff, J., Nagai, H., Reulen, H.-J., Teasdale, G., Eds.; Springer:
Vienna, Austria, 1998; pp. 153–156.
94. Yan, E.B.; Satgunaseelan, L.; Paul, E.; Bye, N.; Nguyen, P.; Agyapomaa, D.; Kossmann, T.; Rosenfeld, J.V.; Morganti-Kossmann, C.
Post-Traumatic Hypoxia Is Associated with Prolonged Cerebral Cytokine Production, Higher Serum Biomarker Levels, and Poor
Outcome in Patients with Severe Traumatic Brain Injury. J. Neurotrauma 2013, 31, 618–629. [CrossRef]
95. Okonkwo, D.O.; Shutter, L.; Moore, C.; Temkin, N.R.; Puccio, A.M.; Madden, C.J.; Andaluz, N.; Chesnut, R.; Bullock, M.R.; Grant,
G.A.; et al. Brain Oxygen Optimization in Severe Traumatic Brain Injury Phase-II. Crit. Care Med. 2017, 45, 1907–1914. [CrossRef]
96. Welch, J.F.; Sutor, T.W.; Vose, A.K.; Perim, R.R.; Fox, E.J.; Mitchell, G.S. Synergy between Acute Intermittent Hypoxia and
Task-Specific Training. Exerc. Sport Sci. Rev. 2020, 48, 125–132. [CrossRef]
97. Gore, A.; Muralidhar, M.; Espey, M.G.; Degenhardt, K.; Mantell, L.L. Hyperoxia sensing: From molecular mechanisms to
significance in disease. J. Immunotoxicol. 2010, 7, 239–254. [CrossRef]
98. Bin-Jaliah, I.; Haffor, A.-S. Ultrastructural Morphological Alterations during Hyperoxia Exposure in Relation to Glutathione
Peroxidase Activity and Free Radicals Productions in the Mitochondria of the Cortical Brain. Int. J. Morphol. 2018, 36, 1310–1315.
[CrossRef]
99. Chong, Z.-Z.; Lin, S.H.; Li, F.; Maiese, K. The Sirtuin Inhibitor Nicotinamide Enhances Neuronal Cell Survival During Acute
Anoxic Injury Through AKT, BAD, PARP, and Mitochondrial Associated. Curr. Neurovascular Res. 2005, 2, 271–285. [CrossRef]
100. Terraneo, L.; Paroni, R.; Bianciardi, P.; Giallongo, T.; Carelli, S.; Gorio, A.; Samaja, M. Brain adaptation to hypoxia and hyperoxia
in mice. Redox Biol. 2017, 11, 12–20. [CrossRef]
101. Felderhoff-Mueser, U.; Bittigau, P.; Sifringer, M.; Jarosz, B.; Korobowicz, E.; Mahler, L.; Piening, T.; Moysich, A.; Grune, T.; Thor, F.;
et al. Oxygen causes cell death in the developing brain. Neurobiol. Dis. 2004, 17, 273–282. [CrossRef]
102. Zhang, Y.; Park, T.S.; Gidday, J.M. Hypoxic preconditioning protects human brain endothelium from ischemic apoptosis by
Akt-dependent survivin activation. Am. J. Physiol. Heart Circ. Physiol. 2007, 292, H2573–H2581. [CrossRef]
103. Nayak, G.H.; Prentice, H.M.; Milton, S.L. Neuroprotective signaling pathways are modulated by adenosine in the anoxia tolerant
turtle. J. Cereb. Blood Flow Metab. 2011, 31, 467–475. [CrossRef]
104. Mao, M.; Zhiling, W.; Hui, Z.; Shengfu, L.; Dan, Y.; Jiping, H. Cellular levels of TrkB and MAPK in the neuroprotective role of
BDNF for embryonic rat cortical neurons against hypoxia in vitro. Int. J. Dev. Neurosci. 2005, 23, 515–521.
105. Digicaylioglu, M.; Bichet, S.; Marti, H.H.; Wenger, R.H.; Rivas, L.A.; Bauer, C.; Gassmann, M. Localization of specific erythropoietin
binding sites in defined areas of the mouse brain. Proc. Natl. Acad. Sci. USA 1995, 92, 3717–3720. [CrossRef] [PubMed]
106. Sanchez, P.E.; Fares, R.P.; Risso, J.-J.; Bonnet, C.; Bouvard, S.; Le-Cavorsin, M.; Georges, B.; Moulin, C.; Belmeguenai, A.; Bodennec,
J.; et al. Optimal neuroprotection by erythropoietin requires elevated expression of its receptor in neurons. Proc. Natl. Acad. Sci.
USA 2009, 106, 9848–9853. [CrossRef] [PubMed]
107. Sakanaka, M.; Wen, T.-C.; Matsuda, S.; Masuda, S.; Morishita, E.; Nagao, M.; Sasaki, R. In vivo evidence that erythropoietin
protects neurons from ischemic damage. Proc. Natl. Acad. Sci. USA 1998, 95, 4635–4640. [CrossRef] [PubMed]
108. Kato, S.; Aoyama, M.; Kakita, H.; Hida, H.; Kato, I.; Ito, T.; Goto, T.; Hussein, M.H.; Sawamoto, K.; Togari, H.; et al. Endogenous
erythropoietin from astrocyte protects the oligodendrocyte precursor cell against hypoxic and reoxygenation injury. J. Neurosci.
Res. 2011, 89, 1566–1574. [CrossRef] [PubMed]
109. Brown, G.C. Nitric oxide and neuronal death. Nitric Oxide 2010, 23, 153–165. [CrossRef]
J. Pers. Med. 2022, 12, 1763 17 of 20

110. Garry, P.S.; Ezra, M.; Rowland, M.J.; Westbrook, J.; Pattinson, K.T.S. The role of the nitric oxide pathway in brain injury and its
treatment—From bench to bedside. Exp. Neurol. 2015, 263, 235–243. [CrossRef]
111. Cheng, A.; Wang, S.; Cai, J.; Rao, M.S.; Mattson, M.P. Nitric oxide acts in a positive feedback loop with BDNF to regulate neural
progenitor cell proliferation and differentiation in the mammalian brain. Dev. Biol. 2003, 258, 319–333. [CrossRef]
112. Li, S.-T.; Pan, J.; Hua, X.-M.; Liu, H.; Shen, S.; Liu, J.-F.; Li, B.; Tao, B.-B.; Ge, X.-L.; Wang, X.-H.; et al. Endothelial Nitric Oxide
Synthase Protects Neurons against Ischemic Injury through Regulation of Brain-Derived Neurotrophic Factor Expression. CNS
Neurosci. Ther. 2014, 20, 154–164. [CrossRef]
113. Brune, B.; Zhou, J. The role of nitric oxide (NO) in stability regulation of hypoxia inducible factor-1α (HIF-1α). Curr. Med. Chem.
2003, 10, 845–855. [CrossRef]
114. Chen, Z.Y.; Wang, L.; Asavaritkrai, P.; Noguchi, C.T. Up-regulation of erythropoietin receptor by nitric oxide mediates hypoxia
preconditioning. J. Neurosci. Res. 2010, 88, 3180–3188. [CrossRef]
115. Kasuno, K.; Takabuchi, S.; Fukuda, K.; Kizaka-Kondoh, S.; Yodoi, J.; Adachi, T.; Semenza, G.L.; Hirota, K. Nitric Oxide Induces
Hypoxia-inducible Factor 1 Activation That Is Dependent on MAPK and Phosphatidylinositol 3-Kinase Signaling. J. Biol. Chem.
2004, 279, 2550–2558. [CrossRef]
116. Haase, V.H. The VHL/HIF oxygen-sensing pathway and its relevance to kidney disease. Kidney Int. 2006, 69, 1302–1307.
[CrossRef]
117. Hoehn, T.; Felderhoff-Mueser, U.; Maschewski, K.; Stadelmann, C.; Sifringer, M.; Bittigau, P.; Koehne, P.; Hoppenz, M.; Obladen,
M.; Bührer, C. Hyperoxia Causes Inducible Nitric Oxide Synthase-Mediated Cellular Damage to the Immature Rat Brain. Pediatr.
Res. 2003, 54, 179–184. [CrossRef]
118. Zhilyaev, S.Y.; Moskvin, A.N.; Platonova, T.F.; Gutsaeva, D.R.; Churilina, I.V.; Demchenko, I.T. Hyperoxic Vasoconstriction in the
Brain Is Mediated by Inactivation of Nitric Oxide by Superoxide Anions. Neurosci. Behav. Physiol. 2003, 33, 783–787. [CrossRef]
119. Rocha-Ferreira, E.; Rudge, B.; Hughes, M.P.; Rahim, A.A.; Hristova, M.; Robertson, N.J. Immediate Remote Ischemic Postcondi-
tioning Reduces Brain Nitrotyrosine Formation in a Piglet Asphyxia Model. Oxidative Med. Cell. Longev. 2016, 2016, 5763743.
[CrossRef]
120. Attaye, I.; Smulders, Y.M.; De Waard, M.C.; Straaten, H.M.O.-V.; Smit, B.; Van Wijhe, M.H.; Musters, R.J.; Koolwijk, P.; Man,
A.M.E.S. The effects of hyperoxia on microvascular endothelial cell proliferation and production of vaso-active substances.
Intensiv. Care Med. Exp. 2017, 5, 22. [CrossRef]
121. Demchenko, I.T.; Oury, T.D.; Crapo, J.D.; Piantadosi, C.A. Regulation of the Brain’s Vascular Responses to Oxygen. Circ. Res.
2002, 91, 1031–1037. [CrossRef]
122. Pham, H.; Vottier, G.; Pansiot, J.; Duong-Quy, S.; Bollen, B.; Dalous, J.; Gallego, J.; Mercier, J.-C.; Dinh-Xuan, A.T.; Bonnin, P.; et al.
Inhaled NO prevents hyperoxia-induced white matter damage in neonatal rats. Exp. Neurol. 2014, 252, 114–123. [CrossRef]
123. Brenner, M.; Stein, D.; Hu, P.; Kufera, J.; Wooford, M.; Scalea, T. Association Between Early Hyperoxia and Worse Outcomes After
Traumatic Brain Injury. Arch. Surg. 2012, 147, 1042–1046. [CrossRef]
124. Davis, W.B.; Rennard, S.I.; Bitterman, P.B.; Crystal, R.G. Pulmonary oxygen toxicity: Early reversible changes in human alveolar
structures induced by hyperoxia. N. Engl. J. Med. 1983, 309, 878–883. [CrossRef] [PubMed]
125. Reynolds, R.A.; Amin, S.N.; Jonathan, S.V.; Tang, A.R.; Lan, M.; Wang, C.; Bastarache, J.A.; Ware, L.B.; Thompson, R.C.
Hyperoxemia and Cerebral Vasospasm in Aneurysmal Subarachnoid Hemorrhage. Neurocritical Care 2021, 35, 30–38. [CrossRef]
[PubMed]
126. Chu, D.K.; Kim, L.H.-Y.; Young, P.J.; Zamiri, N.; Almenawer, S.A.; Jaeschke, R.; Szczeklik, W.; Schünemann, H.J.; Neary, J.D.;
Alhazzani, W. Mortality and morbidity in acutely ill adults treated with liberal versus conservative oxygen therapy (IOTA): A
systematic review and meta-analysis. Lancet 2018, 391, 1693–1705. [CrossRef]
127. Suzuki, S. Oxygen administration for postoperative surgical patients: A narrative review. J. Intensive Care 2020, 8, 79. [CrossRef]
[PubMed]
128. Smit, B.; Smulders, Y.M.; van der Wouden, J.C.; Oudemans-van Straaten, H.M.; Spoelstra-de Man, A.M.E. Hemodynamic effects
of acute hyperoxia: Systematic review and meta-analysis. Crit. Care 2018, 22, 45. [CrossRef] [PubMed]
129. Chatzipanteli, K.; Alonso, O.F.; Kraydieh, S.; Dietrich, W.D. Importance of Posttraumatic Hypothermia and Hyperthermia on the
Inflammatory Response after Fluid Percussion Brain Injury: Biochemical and Immunocytochemical Studies. J. Cereb. Blood Flow
Metab. 2000, 20, 531–542. [CrossRef]
130. Thompson, H.J.; Tkacs, N.C.; Saatman, K.E.; Raghupathi, R.; McIntosh, T.K. Hyperthermia following traumatic brain injury: A
critical evaluation. Neurobiol. Dis. 2003, 12, 163–173. [CrossRef]
131. Truettner, J.S.; Bramlett, H.M.; Dietrich, W.D. Hyperthermia and Mild Traumatic Brain Injury: Effects on Inflammation and the
Cerebral Vasculature. J. Neurotrauma 2017, 35, 940–952. [CrossRef]
132. Huang, T.; Solano, J.; He, D.; Loutfi, M.; Dietrich, W.D.; Kuluz, J.W. Traumatic Injury Activates MAP Kinases in Astrocytes:
Mechanisms of Hypothermia and Hyperthermia. J. Neurotrauma 2009, 26, 1535–1545. [CrossRef]
133. Kurokawa, H.; Ito, H.; Terasaki, M.; Matsui, H. Hyperthermia enhances photodynamic therapy by regulation of HCP1 and
ABCG2 expressions via high level ROS generation. Sci. Rep. 2019, 9, 1638. [CrossRef]
134. Svedung Wettervik, T.M.; Engquist, H.; Lenell, S.; Howells, T.; Hillered, L.; Rostami, E.; Lewén, A.; Enblad, P. Systemic
Hyperthermia in Traumatic Brain Injury—Relation to Intracranial Pressure Dynamics, Cerebral Energy Metabolism, and Clinical
Outcome. J. Neurosurg. Anesthesiol. 2021, 33, 329–336. [CrossRef]
J. Pers. Med. 2022, 12, 1763 18 of 20

135. Saxton, C. Effects of severe heat stress on respiration and metabolic rate in resting man. Aviat. Space Environ. Med. 1981,
52, 281–286.
136. Mickley, G.A.; Cobb, B.L.; Farrell, S.T. Brain hyperthermia alters local cerebral glucose utilization: A comparison of hyperthermic
agents. Int. J. Hyperth. 1997, 13, 99–114. [CrossRef]
137. Nunneley, S.A.; Martin, C.C.; Slauson, J.W.; Hearon, C.M.; Nickerson, L.D.H.; Mason, P.A. Changes in regional cerebral metabolism
during systemic hyperthermia in humans. J. Appl. Physiol. 2002, 92, 846–851. [CrossRef]
138. Spiotta, A.M.; Stiefel, M.F.; Heuer, G.G.; Bloom, S.; Maloney-Wilensky, E.; Yang, W.; Grady, M.S.; Le Roux, P.D. Brain Hyperthermia
After Traumatic Brain Injury Does Not Reduce Brain Oxygen. Neurosurgery 2008, 62, 864–872. [CrossRef]
139. Schumacker, P.T.; Rowland, J.; Saltz, S.; Nelson, D.P.; Wood, L.D. Effects of hyperthermia and hypothermia on oxygen extraction
by tissues during hypovolemia. J. Appl. Physiol. 1987, 63, 1246–1252. [CrossRef]
140. Edvinsson, L.; MacKenzie, E.T.; McCulloch, J. Cerebral Blood Flow and Metabolism; Raven Press: New York, NY, USA, 1993.
141. Crandall, C.G.; Gonzalez-Alonso, J. Cardiovascular function in the heat-stressed human. Acta Physiol. 2010, 199, 407–423.
[CrossRef]
142. Gross, P.M.; Harper, A.M.; Teasdale, G.M. Interaction of histamine with noradrenergic constrictory mechanisms in cat cerebral
arteries and veins. Can. J. Physiol. Pharmacol. 1983, 61, 756–763. [CrossRef]
143. Edvinsson, L. Sympathetic control of cerebral circulation. Trends Neurosci. 1982, 5, 425–429. [CrossRef]
144. Watson, P.; Black, K.E.; Clark, S.C.; Maughan, R.J. Exercise in the heat: Effect of fluid ingestion on blood-brain barrier permeability.
Med. Sci. Sport. Exerc. 2006, 38, 2118–2124. [CrossRef]
145. Watson, P.; Shirreffs, S.M.; Maughan, R.J. Blood-brain barrier integrity may be threatened by exercise in a warm environment.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 288, R1689–R1694. [CrossRef] [PubMed]
146. Walter, E.J.; Hanna-Jumma, S.; Carraretto, M.; Forni, L. The pathophysiological basis and consequences of fever. Crit. Care 2016,
20, 200. [CrossRef] [PubMed]
147. Kilpatrick, M.M.; Lowry, D.W.; Firlik, A.D.; Yonas, H.; Marion, D.W. Hyperthermia in the neurosurgical intensive care unit.
Neurosurgery 2000, 47, 850–856. [CrossRef] [PubMed]
148. Weng, W.-J.; Yang, C.; Huang, X.-J.; Zhang, Y.-M.; Liu, J.-F.; Yao, J.-M.; Zi Zhang, Z.; Wu, X.; Mei, T.; Zhang, C.; et al. Effects of
brain temperature on the outcome of patients with traumatic brain injury: A prospective observational study. J. Neurotrauma
2019, 36, 1168–1174. [CrossRef] [PubMed]
149. Holtzclaw, B.J. The febrile response in critical care: State of the science. Heart Lung J. Crit. Care 1992, 21, 482–501.
150. Bain, A.R.; Smith, K.J.; Lewis, N.C.; Foster, G.E.; Wildfong, K.W.; Willie, C.K.; Hartley, G.L.; Cheung, S.S.; Ainslie, P.N. Regional
changes in brain blood flow during severe passive hyperthermia: Effects of PaCO2 and extracranial blood flow. J. Appl. Physiol.
2013, 115, 653–659. [CrossRef]
151. Stocchetti, N.; Protti, A.; Lattuada, M.; Magnoni, S.; Longhi, L.; Ghisoni, L.; Egidi, M.; Zanier, E. Impact of pyrexia on neurochem-
istry and cerebral oxygenation after acute brain injury. J. Neurol. Neurosurg. Psychiatry 2005, 76, 1135–1139. [CrossRef]
152. Rossi, S.; Zanier, E.R.; Mauri, I.; Columbo, A.; Stocchetti, N. Brain temperature, body core temperature, and intracranial pressure
in acute cerebral damage. J. Neurol. Neurosurg. Psychiatry 2001, 71, 448–454. [CrossRef]
153. Nyholm, L.; Howells, T.; Lewén, A.; Hillered, L.; Enblad, P. The influence of hyperthermia on intracranial pressure, cerebral
oximetry and cerebral metabolism in traumatic brain injury. Upsala J. Med. Sci. 2017, 122, 177–184. [CrossRef]
154. Wang, H.; Wang, B.; Normoyle, K.P.; Jackson, K.; Spitler, K.; Sharrock, M.F.; Miller, C.M.; Best, C.; Llano, D.; Du, R. Brain
temperature and its fundamental properties: A review for clinical neuroscientists. Front. Neurosci. 2014, 8, 307. [CrossRef]
155. Le Roux, P.; Menon, D.K.; Citerio, G.; Vespa, P.; Bader, M.K.; Brophy, G.M.; Diringer, M.N.; Stocchetti, N.; Videtta, W.; Armonda,
R.; et al. Consensus summary statement of the international multidisciplinary consensus conference on multimodality monitoring
in neurocritical care. Neurocritical Care 2014, 21, 1–26. [CrossRef]
156. Rass, V.; Huber, L.; Ianosi, B.-A.; Kofler, M.; Lindner, A.; Picetti, E.; Ortolano, F.; Beer, R.; Rossi, S.; Smielewski, P.; et al. The Effect
of Temperature Increases on Brain Tissue Oxygen Tension in Patients with Traumatic Brain Injury: A Collaborative European
NeuroTrauma Effectiveness Research in Traumatic Brain Injury Substudy. Ther. Hypothermia Temp. Manag. 2020, 11, 122–131.
[CrossRef]
157. Kil, H.Y.; Zhang, J.; Piantadosi, C.A. Brain Temperature Alters Hydroxyl Radical Production during Cerebral Ischemia/Reperfusion
in Rats. J. Cereb. Blood Flow Metab. 1996, 16, 100–106. [CrossRef]
158. Dempsey, R.J.; Combs, D.J.; Maley, M.E.; Cowen, D.E.; Roy, M.W.; Donaldson, D.L. Moderate hypothermia reduces postischemic
edema development and leukotriene production. Neurosurgery 1987, 21, 177–181. [CrossRef]
159. Haba, K.; Ogawa, N.; Mizukawa, K.; Mori, A. Time course of changes in lipid peroxidation, pre-and postsynaptic cholinergic
indices, NMDA receptor binding and neuronal death in the gerbil hippocampus following transient ischemia. Brain Res. 1991,
540, 116–122. [CrossRef]
160. Wells, C.E. The Cerebral Circulation: The Clinical Significance of Current Concepts. Arch. Neurol. 1960, 3, 319–331. [CrossRef]
161. Choi, H.A.; Badjatia, N.; Mayer, S.A. Hypothermia for acute brain injury—mechanisms and practical aspects. Nat. Rev. Neurol.
2012, 8, 214–222. [CrossRef]
162. Dietrich, W.D.; Halley, M.; Valdes, I.; Busto, R. Interrelationships between increased vascular permeability and acute neuronal
damage following temperature-controlled brain ischemia in rats. Acta Neuropathol. 1991, 81, 615–625. [CrossRef]
J. Pers. Med. 2022, 12, 1763 19 of 20

163. Ginsberg, M.D.; Sternau, L.L.; Globus, M.Y.; Dietrich, W.D.; Busto, R. Therapeutic modulation of brain temperature: Relevance to
ischemic brain injury. Cerebrovasc. Brain Metab. Rev. 1992, 4, 189–225.
164. Kramer, R.S.; Sanders, A.P.; Lesage, A.M.; Woodhall, B.; Sealy, W.C. The effect of profound hypothermia on preservation of
cerebral ATP content during circulatory arrest. J. Thorac. Cardiovasc. Surg. 1968, 56, 699–709. [CrossRef]
165. Taft, W.C.; Yang, K.; Dixon, C.E.; Clifton, G.L.; Hayes, R.L. Hypothermia attenuates the loss of hippocampal microtubule-
associated protein 2 (MAP2) following traumatic brain injury. J. Cereb. Blood Flow Metab. 1993, 13, 796–802. [CrossRef] [PubMed]
166. Dede, S.; Deger, Y.; Meral, I. Effect of Short-term Hypothermia on Lipid Peroxidation and Antioxidant Enzyme Activity in Rats. J.
Vet. Med. Ser. A 2002, 49, 286–288. [CrossRef] [PubMed]
167. Truettner, J.S.; Bramlett, H.M.; Dietrich, W.D. Posttraumatic therapeutic hypothermia alters microglial and macrophage polariza-
tion toward a beneficial phenotype. J. Cereb. Blood Flow Metab. 2017, 37, 2952–2962. [CrossRef] [PubMed]
168. Erecinska, M.; Thoresen, M.; Silver, I.A. Effects of Hypothermia on Energy Metabolism in Mammalian Central Nervous System. J.
Cereb. Blood Flow Metab. 2003, 23, 513–530. [CrossRef] [PubMed]
169. Walter, B.; Bauer, R.; Kuhnen, G.; Fritz, H.; Zwiener, U. Coupling of cerebral blood flow and oxygen metabolism in infant pigs
during selective brain hypothermia. J. Cereb. Blood Flow Metab. 2000, 20, 1215–1224. [CrossRef]
170. Jensen, A.; Garnier, Y.; Berger, R. Dynamics of fetal circulatory responses to hypoxia and asphyxia. Eur. J. Obstet. Gynecol. Reprod.
Biol. 1999, 84, 155–172. [CrossRef]
171. Chihara, H.; Blood, A.B.; Hunter, C.J.; Power, G.G. Effect of Mild Hypothermia and Hypoxia on Blood Flow and Oxygen
Consumption of the Fetal Sheep Brain. Pediatr. Res. 2003, 54, 665–671. [CrossRef]
172. Hashem, M.; Zhang, Q.; Wu, Y.; Johnson, T.W.; Dunn, J.F. Using a multimodal near-infrared spectroscopy and MRI to quantify
gray matter metabolic rate for oxygen: A hypothermia validation study. NeuroImage 2020, 206, 116315. [CrossRef]
173. Pichler, G.; Baumgartner, S.; Biermayr, M.; Dempsey, E.; Fuchs, H.; Goos, T.G.; Lista, G.; Lorenz, L.; Karpinski, L.; Mitra, S.; et al.
Cerebral regional tissue Oxygen Saturation to Guide Oxygen Delivery in preterm neonates during immediate transition after
birth (COSGOD III): An investigator-initiated, randomized, multi-center, multi-national, clinical trial on additional cerebral tissue
oxygen saturation monitoring combined with defined treatment guidelines versus standard monitoring and treatment as usual in
premature infants during immediate transition: Study protocol for a randomized controlled trial. Trials 2019, 20, 178. [CrossRef]
174. Laurikkala, J.; Aneman, A.; Peng, A.; Reinikainen, M.; Pham, P.; Jakkula, P.; Hästbacka, J.; Wilkman, E.; Loisa, P.; Toppila, J.;
et al. Association of deranged cerebrovascular reactivity with brain injury following cardiac arrest: A post-hoc analysis of the
COMACARE trial. Crit. Care 2021, 25, 350. [CrossRef]
175. Clifton, G.L.; Allen, S.; Barrodale, P.; Plenger, P.; Berry, J.; Koch, S.; Fletcher, J.; Hayes, R.L.; Choi, S.C. A Phase II Study of Moderate
Hypothermia in Severe Brain Injury. J. Neurotrauma 1993, 10, 263–271. [CrossRef]
176. Rösli, D.; Schnüriger, B.; Candinas, D.; Haltmeier, T. The Impact of Accidental Hypothermia on Mortality in Trauma Patients
Overall and Patients with Traumatic Brain Injury Specifically: A Systematic Review and Meta-Analysis. World J. Surg. 2020,
44, 4106–4117. [CrossRef]
177. Wu, X.; Tao, Y.; Marsons, L.; Dee, P.; Yu, D.; Guan, Y.; Zhou, X. The effectiveness of early prophylactic hypothermia in adult
patients with traumatic brain injury: A systematic review and meta-analysis. Aust. Crit. Care 2021, 34, 83–91. [CrossRef]
178. Tokutomi, T.; Morimoto, K.; Miyagi, T.; Yamaguchi, S.; Ishikawa, K.; Shigemori, M. Optimal Temperature For The Management
Of Severe Traumatic Brain Injury: Effect Of Hypothermia On Intracranial Pressure, Systemic And Intracranial Hemodynamics,
And Metabolism. Neurosurgery 2007, 61 (Suppl. 1), 102–112. [CrossRef]
179. Ghosh, A.; Highton, D.; Kolyva, C.; Tachtsidis, I.; Elwell, C.E.; Smith, M. Hyperoxia results in increased aerobic metabolism
following acute brain injury. J. Cereb. Blood Flow Metab. 2017, 37, 2910–2920. [CrossRef]
180. Yang, D.; Ma, L.; Wang, P.; Yang, D.; Zhang, Y.; Zhao, X.; Lv, J.; Zhang, J.; Zhang, Z.; Gao, F. Normobaric oxygen inhibits AQP4
and NHE1 expression in experimental focal ischemic stroke. Int. J. Mol. Med. 2019, 43, 1193–1202. [CrossRef]
181. Liang, J.; Qi, Z.; Liu, W.; Wang, P.; Shi, W.; Dong, W.; Ji, X.; Luo, Y.; Liu, K.J. Normobaric Hyperoxia Slows Blood–Brain Barrier
Damage and Expands the Therapeutic Time Window for Tissue-Type Plasminogen Activator Treatment in Cerebral Ischemia.
Stroke 2015, 46, 1344–1351. [CrossRef]
182. Braswell, C.; Crowe, D.T. Hyperbaric oxygen therapy. Compend. Contin. Educ. Vet. 2012, 34, E1–E6.
183. Sanchez, E.C. Mechanisms of action of hyperbaric oxygenation in stroke: A review. Crit. Care Nurs. Q. 2013, 36, 290–298.
[CrossRef]
184. Liang, F.; Sun, L.; Yang, J.; Liu, X.-H.; Zhang, J.; Zhu, W.-Q.; Yang, L.; Nan, D. The effect of different atmosphere absolute
hyperbaric oxygen on the expression of extracellular histones after traumatic brain injury in rats. Cell Stress Chaperones 2020,
25, 1013–1024. [CrossRef]
185. Xu, Z.; Huang, Y.; Mao, P.; Zhang, J.; Li, Y. Sepsis and ARDS: The dark side of histones. Mediat. Inflamm. 2015, 2015, 205054.
[CrossRef] [PubMed]
186. Palzur, E.; Vlodavsky, E.; Mulla, H.; Arieli, R.; Feinsod, M.; Soustiel, J.F. Hyperbaric oxygen therapy for reduction of secondary
brain damage in head injury: An animal model of brain contusion. J. Neurotrauma 2004, 21, 41–48. [CrossRef] [PubMed]
187. Palzur, E.; Zaaroor, M.; Vlodavsky, E.; Milman, F.; Soustiel, J.F. Neuroprotective effect of hyperbaric oxygen therapy in brain
injury is mediated by preservation of mitochondrial membrane properties. Brain Res. 2008, 1221, 126–133. [CrossRef] [PubMed]
188. Rockswold, G.L.; Ford, S.E.; Anderson, D.C.; Bergman, T.A.; Sherman, R.E. Results of a prospective randomized trial for treatment
of severely brain-injured patients with hyperbaric oxygen. J. Neurosurg. 1992, 76, 929–934. [CrossRef] [PubMed]
J. Pers. Med. 2022, 12, 1763 20 of 20

189. Rockswold, S.B.; Rockswold, G.L.; Zaun, D.A.; Liu, J. A prospective, randomized Phase II clinical trial to evaluate the effect
of combined hyperbaric and normobaric hyperoxia on cerebral metabolism, intracranial pressure, oxygen toxicity, and clinical
outcome in severe traumatic brain injury. J. Neurosurg. 2013, 118, 1317–1328. [CrossRef]
190. Harch, P.G.; Andrews, S.R.; Rowe, C.J.; Lischka, J.R.; Townsend, M.H.; Yu, Q.; E Mercante, D. Hyperbaric oxygen therapy for
mild traumatic brain injury persistent postconcussion syndrome: A randomized controlled trial. Med. Gas Res. 2020, 10, 8–20.
[CrossRef]
191. Amir, H.; Shai, E. The Hyperoxic-Hypoxic Paradox. Biomolecules 2020, 10, 958. [CrossRef]
192. Annoni, F.; Peluso, L.; Bogossian, E.G.; Creteur, J.; Zanier, E.; Taccone, F. Brain Protection after Anoxic Brain Injury: Is Lactate
Supplementation Helpful? Cells 2021, 10, 1714. [CrossRef]
193. Phillis, J.W.; Song, D.; Guyot, L.L.; O’Regan, M.H. Lactate reduces amino acid release and fuels recovery of function in the
ischemic brain. Neurosci. Lett. 1999, 272, 195–198. [CrossRef]
194. Berthet, C.; Lei, H.; Thevenet, J.; Gruetter, R.; Magistretti, P.J.; Hirt, L. Neuroprotective role of lactate after cerebral ischemia. J.
Cereb. Blood Flow Metab. 2009, 29, 1780–1789. [CrossRef]
195. Horn, T.; Klein, J. Neuroprotective effects of lactate in brain ischemia: Dependence on anesthetic drugs. Neurochem. Int. 2013,
62, 251–257. [CrossRef]
196. Berthet, C.; Castillo, X.; Magistretti, P.J.; Hirt, L. New evidence of neuroprotection by lactate after transient focal cerebral
ischaemia: Extended benefit after intracerebroventricular injection and efficacy of intravenous administration. Cerebrovasc. Dis.
2012, 34, 329–335. [CrossRef]
197. Ichai, C.; Armando, G.; Orban, J.-C.; Berthier, F.; Rami, L.; Samat-Long, C.; Grimaud, D.; Leverve, X. Sodium lactate versus
mannitol in the treatment of intracranial hypertensive episodes in severe traumatic brain-injured patients. Intensiv. Care Med.
2009, 35, 471–479. [CrossRef]
198. Bouzat, P.; Sala, N.; Suys, T.; Zerlauth, J.-B.; Marques-Vidal, P.; Feihl, F.; Bloch, J.; Messerer, M.; Levivier, M.; Meuli, R.; et al.
Cerebral metabolic effects of exogenous lactate supplementation on the injured human brain. Intensiv. Care Med. 2014, 40, 412–421.
[CrossRef]
199. Krep, H.; Breil, M.; Sinn, D.; Hagendorff, A.; Hoeft, A.; Fischer, M. Effects of hypertonic versus isotonic infusion therapy on
regional cerebral blood flow after experimental cardiac arrest cardiopulmonary resuscitation in pigs. Resuscitation 2004, 63, 73–83.
[CrossRef]
200. Ros, J.; Pecinska, N.; Alessandri, B.; Landolt, H.; Fillenz, M. Lactate reduces glutamate-induced neurotoxicity in rat cortex. J.
Neurosci. Res. 2001, 66, 790–794. [CrossRef]
201. Sørensen, A.T.; Ledri, M.; Melis, M.; Nikitidou Ledri, L.; Andersson, M.; Kokaia, M. Altered Chloride Homeo-stasis Decreases the
Action Potential Threshold and Increases Hyperexcitability in Hippocampal Neu-rons. eNeuro 2018, 4. [CrossRef]

You might also like